MXPA98004934A - Diagnosis and treatment of alterations related to aur-1 and / or au - Google Patents

Diagnosis and treatment of alterations related to aur-1 and / or au

Info

Publication number
MXPA98004934A
MXPA98004934A MXPA/A/1998/004934A MX9804934A MXPA98004934A MX PA98004934 A MXPA98004934 A MX PA98004934A MX 9804934 A MX9804934 A MX 9804934A MX PA98004934 A MXPA98004934 A MX PA98004934A
Authority
MX
Mexico
Prior art keywords
aur
polypeptide
seq
further characterized
acid sequence
Prior art date
Application number
MXPA/A/1998/004934A
Other languages
Spanish (es)
Inventor
D Plowman Gregory
G Mossie Kevin
Original Assignee
Sugen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sugen Inc filed Critical Sugen Inc
Publication of MXPA98004934A publication Critical patent/MXPA98004934A/en

Links

Abstract

The present invention relates to AUR-1 and / or AUR-2 polypeptides, nucleic acids encoding said polypeptides to cells, tissues and animals containing nucleic acids, to antibodies to said polypeptides, to analyzes using said polypeptides and to methods that are refer to all of the above, methods are provided to treat, diagnose and select diseases or conditions related to AUR-1 and / or AUR-2, characterized by an abnormal interaction between a polypeptide AUR-1 and / or AUR-2 and a participant of binding of AUR-1 and / or AUR

Description

DIAGNOSIS AND TREATMENT OF ALTERATIONS RELATED TO AUR- 1 Yr ^ Q? UR-2 CAMPO, pg A INVENTION The present invention relates to the novel pro ducine called AURORA and AURORA 2 ("AUR-1 and AUR-2") with the nucleotide sequences encoding AUR-1 and / or AUR-Zr as well as various products and useful methods for the diagnosis and treatment of various diseases and conditions related to AUR-1 and / or AUR-2.
BACKGROUND OF THE INVENTION The following description of the background of the invention is provided to help understand the invention but it is not considered to be prior art with respect to the invention. The transduction of the cellular signal is a fundamental mechanism by which the external stimuli that regulate diverse cellular procedures are retransmitted to the interior of the cells. One of the key biochemical mechanisms in signal transduction involves the reversible phosphorylation of the proteins, which allows the regulation of the activity of mature proteins by altering their structure and function.
The protein kinases best characterized in eukaryotes phosphorylate the proteins in the alcohol moiety in the serine, threonine and tyrosine residues. These kinases are divided into two large groups "those specific for phosphorylating serines and threonines" and those specific for phosphorylating tyrosines. Some kinases, called "double-stranded" kinases, are capable of phosphorylating tyrosine as well as serine / threonine residues. Protein kinases may also be characterized by their location within the cell. Some kinases are proteins of the transmembrane receptor type »able to directly alter their catalytic activity in response to the external environment» for example »to the binding of a ligand. Others are non-receptor-type proteins that lack any transmembrane domain. They can be found in a variety of cellular compartments »from the inner surface of the cell membrane to the nucleus. Many kinases are involved in regulatory cascades where their substrates may include other kinases whose activities are regulated by their phosphorylation process. Finally, the activity of some downstream effector is modulated by the phosphorylation that results from the activation of said path. The family of seri a / threonine kinases includes members that are found at all steps of various signaling cascades. including those that are involved in the control of cell growth »migration, differentiation and ßcrecretion ß phosphorylation of transcription factors that result in altered gene expression» muscle contraction »metabolism of the glucose »the control of cellular protein synthesis and the regulation of the cell cycle.
BRIEF DESCRIPTION OF THE INVENTION The present invention relates to portopeptides AUR-1 and / or AUR-2 »to nucleic acids encoding said polypeptides» to cells containing said nucleic acids »to antibodies to said polypeptides» to analyzes using said polypeptides and to methods that refer to all of the above. The present invention is based on the isolation and characterization of new proteins that have been designated AUR-1 and / or AUR-2. Polypeptides and nucleic acids can be produced using well known and normal synthesis techniques when given the sequences presented herein. AUR-1 and / or AUR-2 are related to serine / treoni kinases with short N-terminal extensions. Drosophila and yeast homologs appear to be involved in mitotic regulation. Human proteins appear to be involved in cancer and / or other alterations in signal transcription. AUR-1 RNA is widely expressed in rapidly dividing cells derived from both normal and tumor tissues. However, "AUR2 RNA is expressed in a more restricted pattern that is low in the more normal tissues or is absent" and is abundant only in a subset of lines of tumor-derived cells, particularly those of local origin. -rectal. Both Auroral and Aurora2 show intermediate expression in the fetal liver, in the testes of adults and in the thymus, suggesting a normal role for these proteins in the meiotic division. Both AUR1 and AUR2 appear to regulate the nuclear division "with altered signaling" which results in polyploid cells. This phenotype is likely due to the erroneous segregation of chromosomes "as seen with its yeast counterpart and IPL1. Given that po iploid is a contrast marker of tumor cells and in defective cells in the tumor suppressor, 53 the role of AUR1 and AUR2 in the transformation of cells is being tested. The primary sequence analysis of human genes reveals that they contain a highly conserved C-terminal protein kinase domain, and a weakly conserved N-terminal domain of "74 to 130 amino acids" that plays a regulatory or regulatory role as a reason for attachment to the substrate. Human genes also contain a cAMP / PKA phosphorylation site. R / KR / KXS / T in the kinase domain activation circuit »suggesting a regulatory path similar to CDC2 / CD related proteins. regulated by the cell cycle. The analysis of Southern »that serves to probe the derivatives of the unique N-terminal region of AUR1 and AUR2. indicates that they exist as single-copy genes in human cells. However, under conditions of low demand, it is possible to detect Sacl fragments of 1.3 Kb and 3.2 kb that weakly hybridize to the AURI probe. The cloning and sequence analysis reveals this region encoding a seudogeno related to AUR1 »that lacks an intron (called AUR3) • with multiple frame shifts. Additionally »immediately upstream of the seudogene AUR3 there is a region with complex inverted repeats, which is predicted to form a very stable fork circuit. The AUR3 DNA sequence is homologous to AUR1 »starting from the first nucleotide of the AUR1 cDNA. Immediately upstream of that site "is the predicted fork circuit of AUR3. The genomic clones of AUR1 are currently being characterized to determine whether the homology with AUR3 continues upstream of this nucleotide and whether the cDNA of AUR1 includes or is preceded by a similar hairpin circuit. The utility of the present invention includes the ability to discriminate inhibitors of cell growth and develop small molecule therapeutics to treat cancers. Thus »in a first aspect» the invention incorporates an isolated »enriched or purified» nucleic acid encoding a polypeptide AUR-1 and / or AUR-2. By "AUR-1 and / or AUR-2 polypeptide" is meant an amino acid sequence substantially similar to that shown in SEQ ID NO: 3 or SEQ ID N0: 4, or fragments thereof. A sequence which is substantially similar will preferably have at least one identity of 9054 (better still) at least 95X and most preferably 99 to 100%). with the sequence of SEQ ID NO: 3 or SEQ ID N0: 4. By "identity" is meant a property of sequences that measures their similarity or their relationship. Identity is measured by dividing the number of identical residues by the total number of residues and by multiplying the product by one hundred. So »two copies of exactly the same sequence would have an entity of 100JÍ. but sequences that are less strongly conserved and have omissions, additions or replacements, may have a lower degree of identity. Those who are experts in the field will recognize that several computer programs are available to determine the identity of sequences. By "isolated" in reference to the nucleic acid, it means a polymer of 6 (preferably 21, better yet 39, and best of all 75) or more conjugated nucleotides "including DNA or RNA" which is isolated from a natural source or one that is synthesized. In certain embodiments of the invention, the longer nucleic acids are preferred, for example, those of 300, 600, 900 or more nucleotides and / or those having at least 50% »60%» 75% »90%, 95% or 99% identity with the full length sequence »shown in SEQ ID NO: i or SEQ ID NO: 2. The isolated nucleic acid of the present invention is unique in the sense that it is not found in the pure or separate state, in nature. The use of the term "isolated" indicates that a sequence that occurs in nature "has been removed from its normal (ie, chromosomal) cellular environment. In such a manner, the sequence can be in a cell-free solution or it can be placed in a different cellular environment. The term does not imply that the sequence is the only chain of nucleotides present, but that it is essentially free (about 90 to 95% purity at least) of non-nucleotide material »naturally associated with it» and so is it is intended to be distinguished from isolated chromosomes. By using the term "enriched" in reference to the nucleic acid "it is meant that the specific DNA or RNA sequence constitutes a significantly larger fraction (2 to 5 times) of the total DNA or RNA present in the cells" or in the solution of interest »that in the normal or diseased cells» or in the cells of the β that took the sequence. This would be caused by a person by preferential reduction in the amount of the other DNA or RNA present, or by preferential increase in the amount of the specific DNA or RNA sequence "or by combination of both. However »it should be noted that" enriched "does not it implies that other DNA or RNA sequences are not present "if not only that the relative amount of the sequence of interest has been significantly increased. The term "significant" is used herein to indicate that the level of increase is useful for the person making said increase "and in general means an increase with respect to other nucleic acids of at least two times" better still. five to ten times »or even more. The term also does not imply that there is no DNA or RNA from other sources. DNA from another source "for example, can comprise yeast or bacterial genome DNA, or a cloning vector" such as PUC19. This term distinguishes itself from events that occur in nature "such as viral infection or tumor-like growths" where the level of a rnRNA can be increased naturally with respect to other mRNA species, that is, the term is intended to cover only those situations in which a person has intervened to raise the proportion of the desired nucleic acid. It is also advantageous for some purposes that the β-nucleotide sequences are in purified form. The term "purified" with reference to the nucleic acid does not require absolute purity (eg, a homogeneous preparation); rather it represents an indication that the sequence is relatively purer than in the natural environment (compared to the natural level, this level should be 2 to 5 times higher, for example, in terms of mg / ml). Individual clones isolated from a cDNA library can be purified to electro-organic homogeneity. The DNA molecules recovered »obtained from these clones» could be obtained directly from total DNA or from total RNA. The cDNA clones do not occur in nature, but rather are obtained preferentially by manipulation of a partially purified substance that occurs in nature (messenger RNA). The construction of a cDNA bank from mRNA involves the creation of a synthetic substance (cDNA) and the individual clones of cDNA can be isolated from this synthetic bank by clonal selection of the cells carried by the cDNA bank. Thus, the procedure that includes the construction of a cDNA bank from mRNA and the isolation of different cDNA clones, results in an approximate purification of 10 * times of the natural message. Thus, the purification of at least an order of magnitude, preferably two or three orders and, better still, of four to five orders of magnitude, is expressly contemplated. By "an AUR-1 and / or AUR-2 polypeptide" is meant 25 (preferably 30 »better yet 35» most preferable 40) or more contiguous amino acids »exposed in the full-length amino acid sequences of SEQ ID NO: 3 or SEQ ID NO: 4 or a functional derivative thereof "as described herein. In certain aspects »polypeptides of 100, 200, 300 or more amino acids are preferred. The AUR-1 and / or AUR-2 polypeptide can be encoded by a full-length nucleic acid sequence or any portion of the full length nucleic acid sequence, as long as a functional activity of the polypeptide is retained. In the preferred embodiments the isolated nucleic acid comprises, consists essentially of or consists of a sequence of nucleic acid designated in the full-length amino acid sequence of SEQ ID NO: 8 or SEQ ID NO: 4, a derivative of the itself »or encodes at least 25» 30 »35» 40 »50, 100» 200 or 300 contiguous amino acids thereof »the polypeptide of AUR-1 and / or AUR-2 comprises» consists essentially of »or consists of at least 25, 30, 35 or 40 contiguous amino acids, of a polypeptide AUR-1 and / or AUR-2. The nucleic acid can be isolated from a natural source by cloning of cDNA or by subtractive hybridization. The natural source can be mammalian (human) blood, semen or tissue and the nucleic acid can be synthesized by the triéster method or by using an automatic DNA synthesizer. In other preferred preferred modalities, the nucleic acid or a unique region is preserved, for example, laß which are useful for the screening of hybridization probes to facilitate the identification and cloning of additional polypeptides, the design of PCR probes to facilitate the cloning of additional polypeptides and obtaining antibodies to polypeptide regions. Examples of amino acid sequences of the present invention include the following amino acid sequences (the isolated, purified or enriched nucleic acids that encode eßtán within the scope of the present invention): ENSYPWPYGRQ (SEQ ID N0: 5) CISGP (SEQ ID N0: 6) QFPO (SEQ ID NO: 7) VNSGQ (SEQ ID N ?: ß) RKEPVTPSA-LV (SEQ ID NO: 9) LMSRSNVQPTAAP (SEQ ID NO: 10) VQNQKQKQLQATSVPH (SEQ ID NO: 11) PVSRPLNNTQK (SEQ ID NO: 12) VMENSSGTPD (SEQ ID NO '13) ILTRHFTID (SEQ ID NO: 14) SKQPLPSAPENNPEEQLASKQK (SEQ ID NO: 15). By "conserved nucleic acid regions" is meant regions present in two or more nucleic acids encoding a polypeptide of AUR-1 and / or AUR-2, to which a particular nucleic acid sequence can hybridize under conditions of low demand Examples of "low requirement conditions" suitable for discriminating polypeptides of AUR-1 and / or AUR-2 encoding the nucleic acid are provided in Abe and coautoreß J. Biol. Chem. »19: 13361 (1992) (incorporated here as reference in the present in its entirety »including any drawing). Preferably »laß conserved regions differ by no more than 5 of 20 nucleotides. By "single nucleic acid region" is meant a sequence present in a full-length nucleic acid encoding an AUR-1 and / or AUR2 polypeptide, which is not present in a sequence encoding any other polypeptide occurring in the nature. Said regions preferably contain 30 to 45 contiguous nucleotides present in the full length nucleotide acid encoding an AUR-1 and / or AUR2 polypeptide. In particular, a single region of nucleic acid is preferably of mammalian origin. The invention also incorporates a nucleic acid probe for the detection of a polypeptide of AUR-1 and / or AUR-2 »or a nucleic acid encoding a polypeptide of AUR-1 and / or AUR-2. in a sample. The nucleic acid probe contains nucleic acid which will flange to a sequence designated in SEQ ID NO: i or SEQ ID NO: 2 or a functional derivative thereof. In preferred embodiments, the nucleic acid probe hybridizes to the nucleic acid encoding at least 12 75, 90 105, 120 150, 200, 250 300 or 350 am contiguous noxae of the full length sequence indicated in SEQ ID N0: io SEQ ID N0: 2 »or a functional derivative thereof. Various low or high requirement hybridization conditions can be used depending on the desired specificity or selectivity. Under demanding hybridization conditions only the highly complementary nucleic acid sequences are hybridized. Preferably, said conditions prevent the hybridization of nucleic acids having one or two inequalities in each 20 contiguous nucleotides. Methods for using the probes include detecting the presence or amount of RNA of AUR-1 and / or AUR-2 in a sample, contacting the sample with a nucleic acid probe »under conditions in which hybridization occurs» and detecting the presence or amount of the probe bound to the RNA of AUR-1 and / or AUR-2. The nucleic acid duplex formed between the probe and a DNA sequence encoding the AUR-1 and / or AUR-2 polypeptide can be used in the identification of the detected nucleic acid sequence (e.g. see Nelson and co-authors in Nonißotropic DNA Probe Techniques, page 275 Academic Prese, San Diego (Kricka editors, 1992), which is incorporated in this way by reference, in its entirety, including any drawing). Equipment can be constructed to carry out said methods, including a container means having a nucleic acid probe disposed therein. The invention also incorporates recombinant nucleic acid, preferably in a cell or in an organism. The recombinant nucleic acid may contain a sequence designated in SEQ ID NO: i or SEQ ID NO: 2 or a functional derivative thereof and a vector or an effective promoter for initiating transcription in a host cell. The recombinant nucleic acid may alternatively contain a "functional" transcription initiation region in a cell »a sequence complementary to an RNA sequence encoding an AUR-1 and / or AUR-2 polypeptide» and a transcriptional termination region »Functional, in a cell. In another aspect, the invention incorporates an isolated, enriched or purified AUR-1 and / or AUR-2 polypeptide. By "isolated", in reference to yet a polypeptide, we mean a polypeptide of 2 (preferably 7, better yet 13, best of all 25) or more, conjugated amino acids, including polypeptides that are isolated from a natural source or that are synthesized. In certain aspects, longer polyptides are preferred, such as those having 402, 407, 413 or 425 contiguous amino acids, designated in SEQ ID N0: 3 or SEQ ID N: 4. The isolated polypeptides of the present invention are unique in the sense that they are not found in the pure or separate state in nature. The use of the term "isolated" indicates that a sequence that occurs in nature has been eliminated from its normal cellular environment. Thus, the sequence can be in a cell-free solution or it can be placed in a different cellular environment. The term does not imply that the sequence is the only chain of amino acids present, but is essentially free (about 90 to 95% purity at least) of non-amino acid material, naturally associated with it. By using the term "enriched" in reference to a polypeptide, it is meant that the specific amino acid sequence constitutes a significantly larger fraction (2 to 5 times) of the total amino acids present in the cells or in the solution of interest, that in normal or diseased cells or in the cells from which the sequence was taken. This could be caused by a person by preferential reduction in the amount of the other amino acids present, or by a preferential increase in the amount of the specific amino acid sequence of interest, or by a combination of both. However, it should be noted that enriched does not imply that other amino acid sequences are not present, but only that the relative amount of the interesting sequence has been significantly increased. The term "significance" is used herein to indicate that the level of increase is useful for the person making said increase, and in general means an increase with respect to the other amino acids of at least double, preferably about 5 to 10 times or even more. The term also does not imply that there are no amino acids from other sources. The amino acid from another source, for example, may comprise the amino acid encoded by a yeast or bacterial genome, or by a cloning vector, such as pUC19. The term is intended to cover only those situations in which man has intervened to raise the proportion of the desired nucleic acid. It is also advantageous for some purposes that a sequence of amino acids be in purified form. The term "purified" in reference to a polypeptide "does not require absolute purity (as a homogeneous preparation)" rather represents an indication that the sequence is relatively purer than in the natural environment (compared to the natural level) this level should be at least 2 to 5 times higher, for example »in terms of rag / ml). The purification of at least an order of magnitude »preferably two or three orders of magnitude and» better still »four to five orders of magnitude» is expressly contemplated. The substance is preferably free from contamination at a functionally significant level, for example, with a purity of 90%, 95% or 99%. In the preferred embodiments »the AUR-1 and / or AUR-2 polypeptide contains at least 25, 30, 35, 40, 50, 100, 150, 200, 250, 300 or 350 contiguous amino acids of the full-length sequence set forth in SEQ ID NO: 3 or SEQ ID NO: or a functional derivative thereof. In yet another aspect, the invention incorporates an antibody (e.g., a monoclonal or polyclonal antibody), which has specific binding affinity to the AUR-1 and / or AUR-2 polypeptide. The antibody contains an amino acid sequence that is capable of specifically binding to a polypeptide of AUR-1 and / or AUR-2. By "specific binding affinity" is meant that the antibody binds to the AUR-1 and / or AUR-2 polypeptides with higher affinity than that which binds to other polypeptides, under specific conditions. Antibodies that have specific binding affinity for the AUR-1 and / or AUR-2 polypeptides can be used in methods for detecting the presence and / or amount of the AUR-1 and / or AUR-2 polypeptide in a sample by contacting the sample with the antibody under conditions such that an immunocomplex is formed and detecting the presence and / or amount of the antibody conjugated to the polypeptide of AUR-1 and / or AUR-2. Diagnostic equipment can be constructed to carry out these methods, which include a first container means containing the antibody and a second container means having a conjugate or a binding partner of the antibody and a marker. In another aspect, the invention incorporates a hybridoma that produces an antibody that has a specific binding affinity to the AUR-1 and / or AUR-2 polypeptide. By "hybridoma" is meant an immortalized cell line, which is capable of secreting an antibody, for example, an antibody to AUR-1 and / or AUR-2. In preferred modes, the antibody for AUR-1 and / or AUR-2 comprises an amino acid sequence that is capable of specifically binding to the AUR-1 and / or AUR-2 polypeptide. In another aspect, the invention describes a polypeptide comprising a recombinant AUR-1 and / or AUR-2 polypeptide or a unique fragment thereof. By "single fragment" is meant an amino acid sequence that is present in the full-length AUR-1 and / or AUR-2 polypeptide, which is not present in any other naturally occurring polypeptide. Preferably, said sequence comprises 6 contiguous amino acids, present in the complete sequence. Better still, said sequence comprises 12 contiguous amino acids present in the complete sequence. Even better still, said sequence comprises 18 contiguous amino acids present in the complete sequence. By "recombinant AUR-1 and / or AUR-2 polypeptide" is meant that it includes a polypeptide produced by recombinant DNA techniques, so that it is distinct from a naturally occurring polypeptide, either at its location (eg, present in a cell or tissue different from that found naturally), in its purity or in its structure. In general, said recombinant polypeptide will be present in a cell, in an amount different from that normally observed in nature. In another aspect, the invention describes a recombinant nucleic acid or cell containing purified nucleic acid encoding a polypeptide of AUR-1 and / or AUR-2. In such cells, the nucleic acid may be under the control of its genomic regulatory elements or may be under the control of exogenous regulatory elements, including an exogenous promoter. By "exogenous" is meant a promoter that is not normally coupled in vivo, transcriptionally, to the coding sequence for the AUR-1 and / or AUR-2 polypeptide.
In another aspect, the invention incorporates a binding agent to the AUR-1 and / or AUR-2 »polypeptide capable of binding to the AUR-1 and / or AUR-2 polypeptide. The binding agent is preferably a purified antibody that recognizes an epitope present in a polypeptide of AUR-1 and / or AUR-2. Other binding agents include molecules that bind to the AUR-1 and / or AUR-2 polypeptide and analogous molecules that bind to an AUR-1 and / or AUR-2 polypeptide. Such binding agents can be identified using assays that measure binding activity of AUR-1 and / or AUR-2, such as those that measure the activity of PDGFR. By "purified" in reference to an antibody, it is meant that the antibody is distinct from the antibody that occurs in nature, such as in purified form. Preferably, the antibody is provided as a homogeneous preparation by common and current techniques. The uses of the antibodies to the cloned polypeptide include those which are to be used as therapeutic tools or as diagnostic tools. The invention incorporates a method for discriminating human cells containing AUR-1 and / or AUR-2 polypeptide or an equivalent sequence. The method incorporates identifying the novel polypeptide in human cells »using techniques that are routine and common in this field» such as those described here to identify AUR-1 and / or AUR-2 (eg »cloning» analysis of Southern or Northern blots »hybridization in if your» PCR amplification »etc.). The invention also incorporates methods for discriminating human cells for AUR-1 and / or AUR-2 polypeptide binding partners. and discrimination from other organisms for AUR-1 and / or AUR-2. or the corresponding union participant. The present invention also incorporates the isolated or enriched purified versions of the peptides identified by the methods described above. In another aspect, the invention provides an analysis to identify agents capable of interfering with the interaction between AUR-1 and / or AUR-2 and a binding partner of AUR-1 and / or AUR-2. Such analyzes can be carried out in vitro or in vivo and are described in detail here or can be obtained by modifying existing analyzes "such as the growth analysis described in the patent application Serial No. 08 / 487,088 filed on June 7, 1995 ( incorporated herein by this reference including any drawings), or the analyzes described in the patent application Serial No. 60 / 005,167, filed on October 13, 1995 ((incorporated herein by reference, including any drawings). could be modified to use the genes of the present invention is described in the international application No. WO 94/23039 »published on October 13, 1994. Other possibilities include detecting the activity of kinase in an autophosphorylation analysis or testing the activity of kinase on common substrates, such as histones, myelin basic protein, gamma-tubulin or protein centers The binding partners can be identified by placing the N-terminal portion of the protein in a two-hybrid screen or by detecting the phosphotyrosine of a double-specificity kinase. Fields and Song, US Patent No. 5,283,183, issued February 1, 1994, which is incorporated herein by reference. A means by which the activity of the Aurora inhibitors can be defined is a discriminating system using a temperature sensitive yeast mutant, such as that described by Chan and Botstein (Genetics 135: 677-691 »1993); see also Francisco and co-authors, Mol. Cell, Bio. 14 (7): 4731-4740, 1994), both incorporated herein by this reference, in its entirety, including any drawings. Briefly, yeast strain CCY72-3D-1 (ipl 1-2), which expresses a temperature-sensitive form of the Aurora yeast homologue (ipil)although it is viable at 26 ° C, it is unable to develop at 37 ° C. Transfection of this strain with an expression plasmid containing a hybrid Aurora gene »containing the N-terminal portion of ipil» containing the domain or interaction domains of putative substrate »and the C-terminal portion of Auroras 1 or 2 »which contains the catalytic domain» is imposed on this sensability at the development temperature. The yeast strain expressing Aurora is then grown at 37 ° C in the presence of a test substance. No development will be evident in the presence of substances that inhibit Aurora's catalytic function. Potential inhibitors include low molecular weight chemicals and / or natural products that are isolated from various organisms such as fungi, marine organisms, plants, etc. The brief description of the invention described above is not limiting and other aspects and advantages of the invention will become apparent from the following description of the preferred embodiments and the claims.
DESCRIPTION OF THE PREFERRED MODALITIES The present invention relates to the polypeptides of AUR-1 and / or AUR-2 »to the nucleic acids encoding said polypeptides, to cells, tissues and animals containing said nucleic acids, to the antibodies to said polypeptides, to the analysis using said polypeptides and methods that relate to all of the above.
I.- THE NUCLEIC ACID THAT CODIFIES POLIPEPTIDES OF AUR-1 AND / 0 ¿UB = 2 Included within the scope of this invention are the functional equivalents of the isolated nucleic acid molecules described herein. The degeneracy of the genetic code allows the replacement of certain codons by other codons that specify the same amino acid and, therefore, that give rise to the same protein. The nucleic acid sequence can vary substantially since »with the exception of methionine and tryptophan» the known amino acids can be encoded by more than one codon. Thus »the portions or the entire AUR-1 and / or AUR-2 gene could be synthesized to give a nucleic acid sequence significantly different from that shown in SEQ ID NO: i or SEQ ID NO: 2. Its encoded amino acid sequence »however, would be retained. Further »the nucleic acid sequence may comprise a nucleotide sequence which is the result of addition» the omission or substitution of at least one nucleotide at the 5r end and / or the 3 'end of the nucleic acid formula shown in SEQ ID NO: 2 SEQ ID NO: 2 or a derivative thereof. Any nucleotide or polynucleotide can be used in this sense "as long as its addition, omission or substitution does not alter the amino acid sequence of SEQ ID NO: 3 or SEQ ID N0: 4, which is encoded by the nucleotide sequence. For example, it is intended that the present invention include any nucleic acid sequence that is the result of the addition of ATG as a start codon at the 5 'end of the nucleic acid sequence of the invention, or its derivative, or the addition of TTA, TA6 or TGA, as a stop codon at the 3T end of the nucleotide sequence of the invention, or its derivative. Ademáß. the nucleic acid molecule of the present invention, as necessary, may have "restriction endonuclease recognition sites" added to its 5 'end and / or its 3T end. Such functional alterations of a given nucleic acid sequence provide an opportunity to promote the secretion and / or processing of heterologous proteins encoded by sequences of foreign amino acids, fused thereto. All variations of the nucleotide sequence of the AUR-1 and / or AUR-2 genes and their fragments allowed by the genetic code "therefore" are included within this invention. Additionally it is possible to omit codons or replace one or more codons with other codons that degenerate the codons to produce a structurally modified polypeptide, but having substantially the same utility or activity of the polypeptide produced by the unmodified nucleic acid molecule. As recognized in the art, the two polypeptides are functionally equivalent, as are the two nucleic acid molecules that give rise to their production, even when the differences between the nucleic acid molecules are not related to the degeneracy of the genetic code .
II.- THE NUCLEIC ACID PROBE FOR THE DETECTION OF AUR-1 AND / OR UR-3 A nucleic acid probe of the present invention can be used to probe an appropriate chromosomal or cDNA bank by "customary hybridization methods" to obtain another nucleic acid molecule of the present invention. A DNA or chromosomal cDNA library can be prepared from appropriate cells »according to recognized methods in this field (see" Molecular Cloning: A Laboratory Manual ", second edition, edited by Sambrook, Fritsch and Maniatis» Cold Spring Harbor Laboratory »1989). Alternatively, chemical synthesis is carried out in order to obtain nucleic acid probes having a β-sequence of nucleotides corresponding to N-terminal and C-terminalleβ portions of the amino acid sequence of the polypeptide of interest. Thus, the synthesized nucleic acid probes can be used as senεi il izareε in a polymerase chain reaction (PCR) carried out according to recognized PCR techniques essentially in accordance with the PCR protocol ß A Guide to Method ß and Appl cations ", edited by Michael and co-authors, Academic Press, 1990, using the appropriate chromosomal or cDNA library to obtain the fragment of the present invention. Those skilled in the art can easily design such probes, based on the sequence described herein, using computer aligning methods and computer sequence analysis, known in the art (see "Molecular Cloning: A Laboratory Manual", second edition). »Edited by Sambrook, Fritsch and Maniatis, Cold Spring Harbor Laboratory, 19B9). The hybridization probes of the present invention can be labeled by common and current labeling techniques, for example, with a radiolabel, an enzyme tag, a fluorescent tag, a biotin-aniin tag, chemiluminescence and the like. After hybridization, the probes can be visualized using known methods. The nucleic acid probes of the present invention include RNA, as well as DNA probes, such as probes that are generated using techniques known in the art. The nucleic acid probe can be immobilized on a solid support. Examples of such solid supports include, but are not limited to, plastics, such as polycarbonate, complex carbohydrates, such as agarose and sepharose, and acrylic resins, such as polyacrylamide and latex granules. The techniques for coupling nucleic acid probes to said solid supports are well known in the art. Suitable test samples for nucleic acid probing methods of the present invention include, for example, cells or nucleic acid extracts of cells or biological fluids. The sample used in the methods described above will vary based on the analysis format »the detection method and the nature of the tissues» the cells or extracts to be analyzed. Methods for preparing the nucleic acid cell extracts are well known in the art and can be easily adapted in order to obtain a sample that is compatible with the method used.
III.- A METHOD THAT IS BASED ON A PROBE AND A DEVICE TO DETECT AUR-A AND / Q AUR-Z A method for detecting the presence of AUR-1 and / or AUR-2 in a sample comprises: (a) contacting the sample with the nucleic acid probe described above »under such conditions that hybridization occurs» and (b) detecting the presence of the probe bound to the nucleic acid molecule. Who is skilled in the art will select the nucleic acid probe according to the techniques known in the art »as described above. Samples to be tested include »but not limited to the as» RNA samples from human tissue. A device for detecting the presence of AUR-1 and / or AUR-2 in a sample comprises at least one container means having the nucleic acid probe described above disposed therein. The kit may further comprise other containers comprising one or more of the following: wash reagents and reagents capable of detecting the presence of the bound nucleic acid probe. Examples of detection reagents include. but without limitation: radio-arched probes »enzymatically labeled probes: horseradish peroxidase, alkaline phosphatase), and affinity-labeled probes (biotin» avidin or esteptavidin). In detail »a device with compartments includes any device in which the reagents are contained in separate containers. Such containers include small glass containers, plastic containers or plastic or paper strips. Such containers allow the efficient transfer of reagents from one compartment to another compartment "so that the samples and reagents are not cross-contaminated" and the agents or solutions of each container can be added quantitatively from one compartment to another. other. Such containers shall include a container accepting the test sample, a container containing the probe or sensitizers used in the analysis, containers containing washing reagents (such as phosphate buffered saline, tris-regulators and the like) and containers containing contain the reactants used to detect the hybridized probe, the bound antibody, the amplified product or the like. One skilled in the art will readily recognize that the nucleic acid probes described in the present invention can be easily incorporated into one of the established equipment formats that are well known in the art.
IV.- DNA CONSTRUCTIONS THAT COMPRISE THE ACID MOLECULE NUCLEIC PE? UR-A AND / 0 AUR-Z AND CELLS CONTAINING SS S CONSTRUCTIONS The present invention also relates to a recombinant DNA molecule comprising »from 5 'to 3'» a promoter effective to initiate transcription in a host cell »and the nucleic acid molecules described above. Furthermore, the present invention relates to a recombinant DNA molecule comprising a vector and the nucleic acid molecules described above. The present invention also relates to a nucleic acid molecule comprising a functional transcription region in a cell »a sequence complementary to an RNA sequence coding for an amino acid sequence corresponding to the previously described polypeptide» and a terminating region of transcription »functional in said cell. The molecules described above may be isolated and / or purified DNA molecules. The present invention also relates to a cell or organism which contains a nucleic acid molecule described above and which is thus capable of expressing a peptide. The polypeptide can be purified from cells that have been altered to express the polypeptide. It is said that an eß cell "altered to express a desired polypeptide" when the cell "by means of genetic manipulation, is forced to produce a protein that normally does not produce or that normally produces the cell at lower levels. Whoever is an expert in the field can easily adapt the procedures to introduce and express genomic sequence?, of cDNA or synthetic »in eukaryotic or prokaryotic cells. It is said that a nucleic acid molecule, such as DNA, is "capable of expressing" a polypeptide, if it contains a nucleotide sequence containing transcriptional and translational regulatory information, and said sequences are "operably linked" to the nucleotide sequences. which encode the polypeptide. An operable link is a link in which the regulatory DNA sequences and the DNA sequence to be expressed are connected in such a way as to allow the expression of the gene sequence. The precise nature of the regulatory regions necessary for the expression of the gene sequence may vary from one organism to another, but will generally include a promoter region that, in the prokaryotes, contains both the promoter (which directs the start of the transcription of RNA), like DNA sequences that, when they are transcribed to RNA, will signal the start of synthesis. Said regions will normally include the sequence ß 5 * -non-coding »involved with the start of transcription and translation» such as the TATA box »the sequence leader» the sequence CAAT and similes. If you want the non-coding region 3T with respect to the sequence encoding the gene AUR-1 and / or AUR-2, it can be obtained by the methods described above. This region can be retained for its transcriptional termination regulatory sequences, such as termination and polyadenylation. Thus »by retaining region 3" naturally contiguous to the DNA sequence encoding an AUR-1 and / or AUR-2 gene, transcription termination signals can be provided When the transcription termination signals are not βßßfactory in the expression of the host cell, then it can be substituted with a functional 3 'region in the host cell.It is said that two DNA sequences (such as a promoter region sequence and a sequence of AUR-1 and / or AUR -2) are operably linked if the nature of the link between the two DNA sequences: (1) does not result in the introduction of a mutation in the frame shift; (2) does not interfere with the ability of the promoter region sequence to direct the transcription of a gene sequence of AUR-1 and / or AUR-2; or (3) does not interfere with the ability of the AUR-1 and / or AUR-2 gene sequence to be transcribed by the promoter region sequence. Thus, a promoter region would be operably linked to a DNA sequence if the promoter were capable of transcribing that DNA sequence. A) Yes. To express an AUR-1 and / or AUR-2 gene, transcriptional and translational signals recognized by an appropriate host are necessary.
The present invention comprises the expression of the AUR-1 and / or AUR-2 gene (or a functional derivative thereof) in prokaryotic or eukaryotic cells. Prokaryotic hosts »in general» are very efficient and convenient for the production of recombinant proteins and »therefore, a type of preferred expression system for the AUR-1 and / or AUR-2 gene. Prokaryotes are very often represented by various strains of E. coli. However, "other microbial strains" can also be used, which include other bacterial strains. In prokaryotic systems, plasmid vectors containing reproduction sites and control sequences derived from a species compatible with the host may be ußadoß. Examples of such suitable plasmid vectors can include pBR322. pUCllβ »pUC119 and the like; suitable phage or bacteriophage vectors which may include ga ma-gtlO ga ma-gtll and the like; and suitable virus vectors which may include pMAM-neo »pKRC and the like. Preferably, the selected vector of the present invention has the capacity to reproduce in the selected host cell. The recognized prokaryotic hosts include bacteria such as E. coli »Bacillus, Streptomyces» Pseudomonas, Salmonella, Serratia and the like. However, under such conditions, the peptide will not be glycosylated. The prokaryotic host must be compatible with the replicon and control sequences in the expression plasmid. To express AUR-1 and / or AUR-2 (or a functional derivative thereof) in a prokaryotic cell, it is necessary to operably link the sequence of AUR-1 and / or AUR-2 to a functional prokaryotic promoter. Said promoters can be constitutive or, even better, regulable (ie, inducible or deprepressible). Examples of constitutive promoters include the int promoter of the bacteriophage lambda, the b a promoter of the beta beta-lactase gene sequence of pBR322, and the CAT promoter of the chloramphenicol-eceti 1 transferase gene sequence of pPR325 and the like. Examples of prokaryotic promoters include the main right and left promoters of bacteriophage lambda (P and PN), the Trp, recA, lambda-acz, lambda-acl and gal promoters of E. coli, alpha-amylase (Ulmanen and co-authors, J. Bacteriol 162: 176-182 (19B5)) and the promoters specific for c-28 from B. subtilis (Gilman and co-authors, Gene sequence 32: 11-20 (1984)), the promoters of bacteriophages from Basilluß (Gryczan , in: The Molecular Biology of the Bacilli, Academic Prese, Inc. NY, USA (1982), and the promoters of Streptomyces (Ward and coauthors, Mol. Gen. Genet., 203: 468-478 (1986)). The prokaryotic promoters are summarized by Glick (J. Ind. Microbiot. 1: 277-282 (1987)); Cenatiempo (Biochimie 68: 505-516 (1986))? and Gottesman (Ann. Rev. Genet .. l?: 415-442 (1984)). Proper expression in a prokaryotic cell also requires the presence of a ribosomal binding site upstream of the gene sequence coding sequence. Said ribosomal binding sites are described, for example, by Gold and coauthors (Ann.Rev. Microbio !. »35: 365-404 (1981)). The selection of the control sequences "of the expression vectors" of the transformation methods and the like depend on the type of host cell used to express the gene. As used herein, "cell", "cell line" and "cell culture" can be used interchangeably and all such designations include progeny. Thus, the words "transformant" or "transformed cells" include the primary subject cell and the cultures derived therefrom, regardless of the number of trans transcepts. It is also understood that all progeny can not be identical in DNA content »due to berated or inadvertent mutations. However, as defined, the mutant progeny has the same functionality as that of the originally transformed cell. Host cells that can be used in the expression systems of the present invention are not strictly limited, as long as they are used in the expression of the peptide of AUR-1 and / or AUR-2 »of interest. Suitable hosts can often include eukaryotic cells. Preferred eukaryotic hosts include, for example, yeast, fungi, i-secto-cells, mammalian cells, either in vivo or in tissue culture. Mammalian cells that can be ußadaß as a hostioneß include HeLa cells, cells of fibroblastic origin, such as VERO or CH0-K1 or cells of lymphoid origin, and their derivatives. Preferred mammalian host cells include SP2 / 0 and J558L, as well as neuroblastoma cell lines, such as IMR 332, which can provide better capabilities to correct post-translational processing. Additionally, plant cells are also available as hosts, and control sequences compatible with the plant cells can be obtained, for example, the 35S and 19S mosaic of cauliflower viruß, and the promoter and polyadenylation signal sequences of nopaline-synthase. Another preferred host is an insect cell »for example» the larvae of Dros? Row. By using insect cells as hosts, the Drosophila alcohol dehydrogenase promoter can be used. Rubín »Science 240: 1453-1459 (19BB). Alternatively, baculovirus vectors can be engineered to express large amounts of AUR-1 and / or AUR-2 in insect cells (Jasny »Science, 238: 1653 (1987) 5 Mi 11er and co-authors, in: Genetic Engineering (1986); Setlow, J.K. and co-authors, editors, Plenu, Volume 8, pages 277-297). Any of a number of yeast gene sequence expression systems, incorporating promoter and terminator elements from actively expressed gene sequences, encoding the glycolytic enzymes, can be used and produced in large amounts. when yeast develops in glucose-rich media. The known neolithic gl gene sequences can also very efficiently provide a transcription control eß signal. The yeast provides substantial advantages in that it can also effect modifications in the peptide after translation. There are several recombinant DNA strategies that use strong promoter sequences and a high number of plasmid copies that can be used for the production of the desired proteins in yeast. The yeast recognizes the leader sequences or the gene products of cloned mammals and secretes peptides carrying the leader sequences (ie, pre-peptides). For a mammal host »various possible vector systems are available for the expression of AUR-1 and / or AUR-2. A wide variety of transcriptional and translational regulatory sequences can be employed, depending on the nature of the host. Transcriptional and translational regulatory signals can be derived from viral sources »such as adenovirus» bovine papilloma viruß »ci to egaloviruß» simian virus or similar »where the regulatory signals are linked to a particular gene sequence, which has a high level of expression. Alternatively, promoters can be used for mammalian expression products, such as actin »collagen» myosin and the like. It is possible to select regulatory signals of the start of transcription that allow repression or activation "so that the expression of the gene sequences can be modulated. Of interest are the regulatory signals that are sensitive to temperature, so that by varying the temperature, expression can be repressed or initiated, or those that are subject to chemical regulation (for example, as a metabolite). The expression of AUR-1 and / or AUR-2 in eukaryotic hosts requires the use of eukaryotic regulatory regions. In general, said regions will include a sufficient promoter region to direct the initiation of RNA synthesis. Preferred eukaryotic promoters include, for example, the promoter of the mouse metallothionein I gene sequence (Ha er and co-authors »J. Mol, APPI, Gen., i: 273-2BB (1982)); the TK promoter of Herpes virus (McKnight. Cell »31: 355-365 (1982)); the SV40 early promoter (Benoist and co-authors »Nature (London) 290: 304-310 (1981)); the yeast gene gal4 sequence promoter (Johnston and co-authors »Proc. Nati, Acad. Sci. (USA) 79: 6971-6975 (1982), Silver and co-authors, Proc. Nati, Acad. Sci. (USA). 81: 5951-5955 (1984)). The translation of the eukaryotic mRNA begins at the codon encoding the first methionine. For this reason it is preferable to ensure that the link between a eukaryotic promoter and a DNA sequence coding for AUR-1 and / or AUR-2 (or a functional derivative thereof) does not contain any intermediate codon that is capable of encoding a methionine (eß say. 3B AUG). The presence of said codons results in a fusion protein formation (if the AUG codon is in the same reading frame as the coding sequence of AUR-1 and / or AUR-2) or a frame shift mutation ( if the AUG codon is not in the same reading frame as the coding sequence of AUR-1 and / or AUR-2). A nucleic acid molecule of AUR-1 and / or AUR-2 and an operably linked promoter can be introduced into a prokaryotic or eukaryotic recipient cell, either as a non-reproductive (or RNA) DNA molecule. which can be a linear molecule or. better still »a closed» covalent »circular molecule. Since these molecules are unable to reproduce autonomously, gene expression can occur through the transient expression of the introduced sequence. Alternatively, permanent expression may occur through integration of the DNA sequence introduced into the host chromosome. A vector that is capable of integrating the desired gene sequences into the chromosome of the host cell can be employed. Cells that have stably integrated the DNA introduced into the chromosomes can be selected by also introducing one or more markers that allow the selection of the host cells containing the expression vector. The label can provide phototrophy to an auxotrophic host »biocide resistance» for example »to antibiotics or heavy metals» such as copper »or the like. The selectable marker gene sequence may also be linked directly to the DNA gene sequences to be expressed or introduced into the same cell by co-transfection. It is also necessary that there be additional elements for the optimal synthesis of the single chain link protein mRNA. These elements may include splicing signals as well as transcription promoters »enhancers and termination signals. The cDNA expression vectors incorporating said elements include those described by Okayama »Molec. Cell. Biol. 3: 280 (1983)). The introduced nucleic acid molecule can be incorporated into a plasmid or viral vector »capable of autonomous reproduction in a recipient host. Any of a large number of vectors can be used for that purpose. Important factors in selecting a particular plasmid or viral vector include: the ease with which recipient cells that contain the vector can be recognized and selected from recipient cells that do not contain the vector; the number of copies of the vector desired in a particular host "and whether it is convenient to be able to" exchange "the vector between host cells of different species. Preferred prokaryotic vectors include plasmids, such as those capable of reproduction in E. coli (such as, for example, pBR322 »ColEl» pSClOl »pACYC 184, nVX.These plasmids, for example, are described by Sambrook (cf. "Molecular Cloning: A Laboratory Manual", second edition, edited by Sambrook, Fritßch and Maniatis, Cold Spring Harbor Laboratory, 1989). Bacillus plasmids include pC194. pC221, pT127 and the like. Such plasmids are described by Gryczan (in The Molecular Biology of the Bacilli, Academic Press, Inc. NY, USA (1982), pages 307-329). Suitable streptomyces plasmids include pIJIOl (Kendall and co-authors, J. Bacteriorl., 169: 4177-4183 (19B7)) and streptomyces bacteriophages. such as TC31 (Chater and co-authors, in: Sixth International Symposium on Actino ycetales Biology, Akademiai Kaido, Budapest, Hungary (1986), pages 45-54). The pseudomonas plasmids are summarized by John and co-authors (Rev. Infect. Dis., 8: 693-704 (19B6)) and by Izaki (Jpn. J. Bacteriol., 33: 729-742 (1978)). Preferred eukaryotic plasmids include, for example, BPV »vaccines, SV40, circle of 2 microns, and the like, or their derivatives. Such plasmids are well known in the art (Bolstein and co-authors, Miami Wntr Sympo, 19: 265-274 (1982); Broach in: "The Molecular Biology of the Yeast Saccharomyces: Life Circle and Inheritance", Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA pages 445-470 (1981); Broach, Cell, 28: 203-204 (1982); Bollón and co-authors J. Ctin. Hematol. Oncol. 10: 39-48 (1980); Maniatis, in: Cell Biology: A Comprehenßi ve Treatiße, Volume 3, Gene Sequence Expreßßion, Academic Press, NY, USA pages 563-608 (1980).
Once the vector or nucleic acid molecule containing the construction or constructs for expression has been pred, the DNA construct or constructs can be introduced into the appropriate host cell, by any of a variety of suitable means »That is to say» transformation »transfection» conjugation »fusion of the protoplast. electroporation particle gun technology »precipitation with calcium phosphate» direct injection meroinjection and the like. After the introduction of the vector, the recipient cells are developed in a selective medium, which selects the development of the cells containing the vector. The expression of the cloned molecule or gene molecules results in the production of AUR-1 and / or AUR-2 or fragments thereof. This can take place in the transformed cells as such, or after the induction of these cells to differentiate (for example, by administration of bromodeoxyurea to neuroblastoma cells or the like). A variety of incubation conditions can be used to form the peptide of the present invention. The most preferred conditions are those in which the medium mimics physiological conditions.
V.- POLYPEPTIDES OF AUR-1 AND / OR AUR-2 PURIFIED A variety of methodologies known in the art can be used to obtain the peptide of the present invention. The peptide can be purified from tissues or cells that naturally produce the peptide. Alternatively, "isolated nucleic acid fragments" described above could be used to express the AUR-1 and / or AUR-2 protein in any organism. Samples of the present invention include cells, protein extracts or cell membrane extracts, or biological fluids. The sample will vary based on the analysis format, the detection method and the nature of the tissues, cells or extracts used as samples. Any eukaryotic organism can be used as a source for the peptide of the invention, as long as the source organism contains said peptide. As used herein, "source organism or source organism" refers to the original organism from which the amino acid sequence of the subunit is derived, regardless of the organism in which the subunit is expressed and finally isolated. Whoever is an expert in the matter will easily follow the methods to isolate proteins, in order to obtain the peptide free of natural contaminants. These include, but are not limited to: size exclusion chromatography, HPLC, ion exchange chromatography, and chromatography by one affinity.
VIt- AN ANTIBODY THAT HAS AFINIPAP PE UNION TO A PQLIPEPTIPQ OF AUR-l AND / OR AUR-2. AND A HYBRIDOMA THAT CONTAINS THE ANTIBODY The present invention relates to an antibody having binding affinity to an antibody of AUR-1 and / or AUR-2. The polypeptide may have the amino acid sequence set forth in SEQ ID NO: tt or SEQ ID NO: 4, or a functional derivative thereof, or at least 9 contiguous amino acids thereof (preferably »at least 20, 30 »35 or 40 of its contiguous amino acids). The present invention also relates to an antibody that has specific binding affinity to a polypeptide of AUR-1 and / or AUR-2. Said antibody can be isolated by comparing its binding affinity to an AUR-1 and / or AUR-2 polypeptide with its binding affinity to another polypeptide. Those that bind selectively to AUR-1 and / or AUR-2 will be selected to be used in methods that require a distinction between AUR-1 and / or AUR-2 and other polypeptides. Such methods could include, but are not limited to, analysis of altered expression of AUR-1 and / or AUR-2 in tissue that contains other polypeptides. The AUR-1 and / or AUR-2 proteins of the present invention can be used in a variety of methods and methods, for example, for the generation of antibodies, for use in the identification of pharmaceutical compositions and for studying the interaction of DNA / protein.
The peptide of AUR-1 and / or AUR-2 of the present invention can be used to produce antibodies or hybridomas. Whoever is skilled in the art will recognize that if an antibody is desired, said peptide will generate as described herein and be used as an immunogen. The antibodies of the present invention include monoclonal and polyclonal antibodies. as well as fragments of these antibodies and humanized forms. The humanized forms of the antibodies of the present invention can be generated using one of the methods known in the art, such as chimerization or grafting of RDC. The present invention also relates to a hybridoma that produces the monoclonal antibody described above, or a binding fragment thereof. A hybridoma is an immortalized cell line that is capable of secreting a specific monoclonal antibody. In general, the techniques for preparing monoclonal antibodies and hybridomas are well known in the art (Campbell, "Monoclonal Antibody Technology: Laboratory Techniques in Biochemistry and Molecular Biology" »Elsevier Science Publishers, Amsterda, Loe Pais Netherlands (1984); and co-authors, J. Immunol, Methods 35: 1-21 (19B0)). Any animal (mouse, rabbit and the like) that is known to produce antibodies, can be immunized with the polypeptide released. Methods for immunization are well known in the art. The methods include subcutaneous or intraperitoneal injection of the polypeptide. One of skill in the art will recognize that the amount of polypeptide used for immunization will vary based on the animal being immunized, on the antigenicity of the polypeptide and at the site of the injection. The polypeptide can be modified or an adjuvant can be administered in order to increase the antigenicity of the peptide. Methods for increasing the antigenicity of a polypeptide are well known in the art. Such methods include coupling the antigen with a heterologous protein (such as globulin or β-galactosidase) or by means of the addition of an adjuvant during immunization. For monoclonal antibodies, the spleen cells of the immunized animals are removed, fused with myeloma cells, such as myelone cells SP2 / 0-Agl4 and allowed to become hybridoma cells producing monoclonal antibody. Any of a number of methods well known in the art can be used to identify the hybridoma cells that produce an antibody with the desired characteristics. These include the selection of hybridoma with an ELISA, Western blot analysis or radioimmunoanalysis (Lutz and coautoreß, EXP, Ce11 Res. 175: 109-124 (1988)). Hybridomas secreting the desired antibodies are cloned and the claße and subclass are determined using procedures known in the art ("Monoclonal Antibody Technology: Laboratory Techniques in Biochemistry and Molecular Biology", supra (1984)). For polyclonal antibodies »to the antisera containing antibody from the immunized animals and the presence of antibodies with the desired specificity is discriminated» using one of the procedures described above. The antibodies described above can be detectably labeled. Antibodies can be detectably labeled by the use of radioisotopes »affinity tags (such as biotin» avidin and the like) »enzymatic tags (such as horseradish peroxidase» alkaline phosphatase and the like) »fluorescent tags (such as FITC or rhodamine and the like), atoms for agnostics. and similar. Methods for obtaining such labeling are well known in the art, for example, see (Stemberger and co-authors »J. Histochem, Cytochem.» 18: 315 (1970), Bayer and co-authors, Met. Enzym. 62: 308 (1979). Engval and coauthors, Immunot 109: 129 (1972) 5 Goding, J. Im unol.Meth. 13: 215 (1976)). The labeled antibodies of the present invention can be used for in vitro analysis in vitro or in situ to identify cells or tissues that express a specific peptide. It is also possible to immobilize the antibodies described above on a solid support. Examples of such solid supports include plastics, such as polycarbonate, complex carbohydrates such as agarose and sepharose, acrylic resins such as polyacrylamide and latex granules. The techniques for coupling antibodies to said solid supports are well known in the art.
(Weir and co-authors »" Handbook of Experimental Imology ", 4th edition» Blackwell Scientific Publ cat onß »Oxford, England» Chapter 10 (19B6), Jacoby and co-authors, Meth. Enzym. 34 »Academic Press NY» USA (1974 )). The immobilized antibodies of the present invention can be used for in vivo analysis "in vitro and in situ" as well as in micro-photomyography. On the other hand, one of ordinary skill in the art can readily adapt the currently available methods, as well as the techniques, methods and equipment described above with respect to antibodies to generate peptides capable of binding to a specific peptide sequence. In order to generate the rationally designed antipeptide peptides, for example, see Hurby and coautoreß, "Application of Synthetic Peptides: Antisenße Peptides", in Synthetic Peptides, A User's Guide, WH Freeman, NY, USA, pages 289-307 (1992) and Kaspczak and coauthors, Biochemistry 2B: 9230-B (1989). The peptides can be generated in the peptide by replacing the basic amino acid residues in the peptide sequence of AUR-1 and / or AUR-2 with residue β acids, while keeping the polar groups hydrophobic and uncharged. For example, the residue is replaced lysine »arginine and / or histidine by a-beta-glutamic acid and glutamic acid residues are replaced by lysine, arginine or histidine.
VII.- A METHOD AND A TEAM THAT ARE BASED ON THE ANTIBODY. TO DETECT AUR-1 AND / OR AUR-2 The present invention comprises a method for detecting a polypeptide of AUR-1 and / or AUR-2 in a sample, comprising: (a) contacting the mueßtra with the antibody described above. under conditions such that they form immunocomplexes; and (b) detecting the presence of the antibody bound to the polypeptide. In detail, the methods comprise incubating a test sample with one or more of the antibodies of the present invention and analyzing whether the antibody binds to the test sample. The altered levels of AUR-1 and / or AUR-2 in a mueßtra. compared to normal levels, they may indicate illness. The conditions for incubating an antibody with a test sample vary. The incubation conditions depend on the format used in the analyzes, the detection methods used and the type and nature of the antibody used in the analysis. Those skilled in the art will recognize that any of the currently available immunological analysis formats (such as radioimmunoassays, enzyme-linked immunosorbent assays, Oucheterlony-based diffusion or rocket immunofluorescence assays) can be easily adapted for employ the antibodies of the present invention. Examples of such analyzes may be found in Chard, "An Introduction to Radioimmunoassay and Related Technique" Elsevier Science Publishers, Amsterdam, Loe The Netherlands (1986); Bul lock and coauthors, "Techniques in Immunocytochemistry", Academic Prese, Orlando Florida »USA» Vol. 1 (1982), Vol. 2 (19B3), Vol. 3 (1985) 5 Tijsßen, "Practice and Theory of Enzyme Immunoassays: Laboratory Technique in Biochemistry and Molecular Biology". Elsevier Science Publishers, Amsterdam, The Netherlands (1985). The test samples in the immunological analysis of the present invention include cells, protein extract or membrane of the cells or biological fluids such as blood, serum, asthma or urine. The test sample used in the method described above will vary based on the format of the analysis, on the nature of the detection method and on the tissues, cells or extract ß used as the sample to be analyzed. Methods for preparing protein extracts or cell membrane extracts are well known in the art and can be easily adapted in order to obtain a sample that is capable with the system used. A kit contains all the necessary reagents to carry out the detection methods described previously. The kit may comprise: (i) a first container medium containing an antibody described above; and (ii) a second container means containing a conjugate comprising a binding partner of the antibody and a marker. In another preferred embodiment, the kit further comprises one or more different containers comprising one or more of the following: wash reagents and reagents capable of detecting the presence of bound antibodies. Examples of detection reagents include, but are not limited to, labeled secondary antibodies or, ternetically, if the primary antibody is labeled, chromophore, enzyme or antibody binding reagents, which are capable of reacting with the labeled antibody. . The equipment with compartments can be as described above for nucleic acid probe kits. Those skilled in the art will readily recognize that the antibodies described in the present invention can be easily incorporated into one of the established equipment formats, which are well known in the art.
VIII.- INSULATION OF THE COMPOUNDS THAT INTERACT WITH AUR-1 AND / Q AUR-Z The present invention also relates to a method for detecting a compound capable of binding to a polypeptide of AUR-1 and / or AUR-2, which comprises incubating the compound with AUR-1 and / or AUR-2 and detecting the presence of the compueßto joined to AUR-1 and / or AUR-2. The compound may be present within a complex mixture »eg» fluid »serum of the body or extracts of cells. The present invention also relates to a method for detecting an agonist or antagonist of the activity of AUR-1 and / or AUR-2, or of the activity of binding partner of AUR-1 and / or AUR-2 »comprising incubating cells that produce AUR-1 and / or AUR-2 in the presence of a compound and detect changes in the level of AUR-1 and / or AUR-2 activity or the binding partner activity of AUR-1 and / or AUR-2. The compounds thus identified would produce a change in activity "which would indicate the presence of the compound. The compound may be present within a complex mixture »eg» fluid »serum of the body or extracts of cells. Once the compound is identified it can be isolated using techniques well known in the art. The present invention also comprises a method for agonizing (stimulating) or antagonizing the activity associated with AUR-1 and / or AUR-2 in a mammal »comprising administering to the mammal an agonist or antagonist for AUR-1 and / or AUR- 2 »in an amount sufficient to effect said agonism or antagonism. A method for treating the diseases in a mammal with an agonist or antagonist of the activity related to AUR-1 and / or AUR-2 »comprises administering the agonist or antagonist to a mammal in an amount sufficient to agonize or antagonize the aßocydase functions with AUR-1 and / or AUR-2 »and also included in the previous request.
IX.- TRANS-ANIMAL ANIMALS A variety of methods are available for the production of transgenic animals associated with the present invention. DNA can be injected into the pronucleus of a fertilized egg before the fusion of the male and female pronuclei »or it can be injected into the nucleus of an embryonic cell (for example» in the nucleus of a two-cell embryo) »after of the start of cell division (Brinster and coauthors, Proc. Nati, Acad. Science, USA, 82: 4438-4442 (1985)). It can infect embryos, especially retroviruses. modified to carry the inorganic ion receptor nucleotide sequences of the present invention. Cells of pluripotent origin, derived from a mass of internal cells of the embryo and eßtabi 1 raised in culture, can be manipulated in a culture to incorporate the nucleotide sequences of the present invention. A transgenic animal can be produced from these cells by implantation in a bastocyte implanted in an adoptive mother and allowed to come to term. It is encouraged that suitable transgenic experiments can be obtained from normal commercial sources such as Charles River (Wilmington »MA» USA »Taconic (Germantown» NY, USA). They would make Sprague Dawley (Indianapolis, USA). etc. The procedures for the manipulation of the rodent embryo and for the ieroinjection of DNA into the pronucleus of the zi oto are well known to those of ordinary skill in the art (Hogan and co-authors, supra). Microinjection procedures for fish, amphibian and bird eggs are detailed in Houdebine and Chourrout, Experientia 47: 897-905 (1991). Other methods for introducing DNA into animal tissues are described in U.S. Patent No. 4,945,050 (Sandford and co-inventors, July 30, 1990). By way of example only, to prepare a transgenic mouse. it is induced to surpass uTen the female mice. The females are placed with the males and the mated females are sacrificed by means of asphyxiation with C03 or by cervical dislocation, and loe embryos of oviducts excised. The surrounding cumuluß cells are removed. Then the pronuclear embryos are washed and stored until the moment of injection. Pairs of adult female mice »with random cycles» are formed with vasectomized males. The receiving females are mated at the same time as the donor females. The embryos are then transferred surgically. The procedure to generate transgenic rat eß similar to that of mice. See Ham er and co-authors. Cell. 63: 1099-1112 (1990). Methods for the culture of embryonic stem cells (ES) and the subsequent production of transgenic animals by introducing DNA into ES cells "using methods such as electroporation» precipitation with calcium phosphate / DNA and by direct injection, they are also well known to ordinary experts in the field, see, for example. Teratocarcino as and Embtyonic Stem Cells. A Practical Approach. E.J. Robertson. editor. IRL Press (1987). In the case involving the random integration of genes, a clone containing the sequence or sequences of the invention is transfected with a gene encoding resistance. Alternatively, the gene encoding the neomycin resistance is physically linked to the sequence or sequences of the invention. Transfection and aging of the desired clones is carried out by any of several methods well known to those of ordinary skill in the art (E. Robertson, supra). DNA molecules introduced into ES cells can also be integrated into the chromosome by the homologous recombination procedure. Capecchi »Science» 244: 1288-1292 (1989). Methods for the positive selection of the recombination event (ie say the neo-resistance) and double positive-negative selection (ie, the neo-resistance and resistance to ganciclovir) and the sub-sequential identification of the desired clones by PCR . they have been described by Capecchi. supra and Joyner and co-authors »Nature. 338: 153-156 (19B9). whose teachings are incorporated here. The final phase of the procedure is to inject the target ES cells into blastocysts and transfer the blastocysts to prendeudopreñadaß females. The resulting chimeric animals are bred and the pups are analyzed by Southern blotting to identify the individuals carrying the transgene. Procedures for the production of non-rodent mammals and other animals have been discussed by others. See Houdebine and Chourrout. supra; Pursel and Coauthors Seience »244: 1281-1288 (1989); and Simms and co-authors »Bio / Technology 6: 179-183 (19BB). Thus »the invention provides transgenic non-human mammals containing a transgene encoding a polypeptide of AUR-1 and / or AUR-2» or a gene that effects the expression of a polypeptide of AUR-1 and / or AUR -2. Said transgenic non-human mammals are particularly useful as an in vivo test system to study the effects of introducing a polypeptide of AUR-1 and / or AUR-2 »to regulate the expression of AUR-1 and / or AUR-2 (i.e. »By the introduction of additional genes» nucleic acids of opposite direction »or ribosomes). A "transgenic animal" is an animal that has cells that contain DNA that has been artificially grafted into a cell "and that DNA becomes part of the genome of that animal that develops from that cell. The transgenic animals preferß ßon pri atee »ratoneß» rats »cows» pigs »horses» goats »sheep» dogs and cats. The transgenic DNA can encode a human AUR-1 and / or AUR-2 polypeptide. The natural expression in an animal can be reduced by providing an amount of RNA or DNA of opposite sense effective to reduce the expression of the receptor.
X.- THERAPY WITH GENES AUR-1 and / or AUR-2 or its genetic sequences may also be useful in gene therapy (summarized in My 11th Nature, 357: 455-460 (1992).) My 11th notes that advances have resulted practical approaches in therapy with human genes, which have demonstrated positive initial results.The basic science of gene therapy is described in Mulligan, Science, 260: 926-931 (1993) .In a preferred embodiment, an expression vector that contains the coding sequence of AUR-1 and / or AUR-2 eß inserted into the cells, the cells are developed in vitro and then infused into large numbers of patients, in another preferred embodiment, a segment of DNA containing a selection promoter (eg, a strong promoter) in cells containing an endogenous AUR-1 and / or AUR-2, such that the promoter segment increases the expression of the endogenous AUR-1 and / or AUR-2 gene ( for example, ß transfers the promoter segment to the cell of such ma nera that is directly linked to the gene of AUR-1 and / or endogenous AUR-2). Gene therapy may involve the use of an adenovirus containing the cDNA of AUR-1 and / or AUR-2, to which it targets a tumor, systemic increase of AUR-1 and / or AUR-2 by cell implantation engineered, injection with AUR-1 and / or AUR-2 virus or DNA injection of natural AUR-1 and / or AUR-2, into appropriate tissues. The populations of target cells can be modified by introducing altered forms of one or more of the components of the protein complexes, in order to modulate the activity of said complexes. For example, by reducing or inhibiting the activity of a complex component within the target cells, one or more abnormal signal transduction events, which lead to a condition, can be decreased, inhibited or reversed. One-component, by default, or missense mutants, which retain the ability to interact with other components of protein complexes, but can not function in signal transduction, can be used to inhibit a damaging signal transduction event abnormal Expression vectors derived from viruses, such as retroviruses, vaccinia viruses, adenoviruses, adeno-associated viruses, herpes viruses, various RNA viruses or bovine papilloma viruß, may be used for the delivery of nucleotide sequences (e.g., cDNA) ), which encode the recombinant protein of AUR-1 and / or AUR-2 »in the population of target cells (eg, tumor cells). Methods that are well known to those skilled in the art can be used to construct recombinant viral vectors containing coding sequences. see »for example» the techniques described in Maniatis and coauthors »Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory» NY »USA (1989) and in Ausubel and coauthors» Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, NY, USA (1989). Alternatively 'laß recombinant nucleic acid molecules encoding protein sequences can be used as natural DNA or in a reconstituted system' eg 'liposomes or other lipid systems' for delivery to target cells (eg »see Felgner and coauthors» Nature »337: 3B7-B (1989)). Various other methods for direct transfer of plasmid DNA to cells exist for use in therapy with human genes and involve targeting DNA to receptors in cells by complexing plasmid DNA with proteins. See My 11er »above. In its simplest form, gene transfer can be carried out by simply injecting tiny amounts of DNA into the nucleus of a cell through a micro-ejection procedure. Capecchi, MR. Cell 22; 479-88 (1980). Once the recombinant genes are introduced into a cell, they can be recognized by the normal mechanisms of the cells for transcription and translation, and a gene product will be expressed. Other methods have also been tried to introduce DNA into larger numbers of cells. These methods include transfection. where the DNA is precipitated with CaPO ^ and collected in cells by pinocytosis (Chen C. and Okayama H.
Mol. Cell Biol. 7: 2745-52 (19B7)) 5 electroporation. wherein the cells are exposed to large pulses of voltage to introduce holes in the membrane (Chu G and coauthors, Nucleic Acids Res. 15: 1311-26 (1987)); 1 ipofection / liposome fusion, where the DNA is packed into lipophilic vesicles that fuse with a target cell (Felgner PL and coautoreß, Proc Nati Acad Sci USA 84: 7413-7 (1987)); and particle bombardment, using DNA attached to small projectiles (Yang NS and coauthors, Proc Nati Acad Sci 87: 9568-72 (1990)). Another method for introducing DNA into cells is to attach the DNA to chemically modified proteins. It has also been shown that the adenovirus proteins are able to destabilize the endosomes and increase the absorption of DNA in the cells. The mixture of adenovirus with solutions containing DNA complexes or the binding of DNA to polysilicon covalently bound to the adenovirus using protein crosslinking agents, substantially improves the collection and expression of the recombinant gene. Curiel DT and co-authors, Am. J. Respir. Cell Mol. Biol., 6: 247-52 (1992). As used herein, "gene transfer" means the process for introducing a foreign nucleic acid molecule into a cell. Gene transfer is commonly carried out to increase the expression of a particular product encoded by the gene. The product can include a protein »a polypeptide» a DNA or RNA of opposite sense or an enzymatically active RNA. The transfer of genes into cultured cells or by direct administration to animals can be carried out. Gene transfer generally involves the procedure of contacting the nucleic acid with a target cell through nonspecific or receptor-mediated interactions »the reception of nucleic acid in the cell by means of the membrane or by endocytosis» and the release of the nucleic acid in the cytoplasm from the plasma »of the membrane or the endosome. The expression may also require »the movement of the nucleic acid towards the nucleus of the cell and the binding to appropriate nuclear factors» for transcription. As used in the present "gene therapy" it is a form of gene transfer and is included within the definition of gene transfer "such as ßε ußa in the present" and refers specifically to the transfer of genes to express a therapeutic product from a cell in vivo or in vitro. Transference of ex vivo genes into cells that are then transplanted into a patient can be carried out or carried out by direct adinistration of the nucleic acid or the nucleic acid-protein complex in the patient. In another preferred embodiment, "a vector is provided which has nucleic acid sequences encoding an AUR-1 and / or AUR-2" wherein the nucleic acid sequence is expressed only in specific tissue. Methods for obtaining tissue-specific gene expression "are set forth in International Publication No. WO 93/09236" filed on November 3, 1992 and published May 13, 1993. In all preceding vectors "indicated above" Another aspect of the invention is that the nucleic acid sequence contained in the vector may include additions »omissions or modifications to some or all of the nucleic acid sequence» as defined above. In another preferred modality "a method for the replacement of genes is indicated. "Gene replacement" as used herein means supplying a nucleic acid sequence that is capable of being expressed in vivo in an animal and thereby providing or increasing the function of an endogenous gene that is either defective or defective. in the animal.
EXAMPLES The following examples are non-limiting and are only representative of the various aspects and characteristics of the present invention. The following examples demonstrate the isolation and characterization of the novel proteins AUR-1 and AUR-2. Protein kinases are one of the largest families of eukaryotic proteins with several hundred known members. These proteins share a domain of 250 to 300 amino acids »that can be subdivided into 12 different sub-manifolds that comprise the common catalytic core structure. These conserved protein motifs have recently been exploited using PCR-based cloning strategies, which lead to a significant expansion of known kinases. The multiple alignment of the sequences in the catalytic domain of the protein kinases and the subsequent phylogenetic analysis allows their segregation to a phylogenetic tree. Thus »related kinases are grouped into distinct branches or subfamilies that include: tyrosine kinases, kinases that depend on the cyclic nucleotide» calcium / calmodulin kinases, cyclin-dependent kinases and MAP-qui asaß, as well as several other subfamilies less defined. Initially we proceed to identify the homologs of CCK4 »a receptor that represents a different family of tyrosine kinases. Multiple alignments suggested that CCK4 was more closely related to REOS and to the TRK family of receptor tyrosine kinases. Degenerate sensitizers were designed to preserve the sequences within the kinase I and IX subdomains of those receptors. Subdomain I is at the N-terminus of the kinase domain and contains the consensus motif GXGXXGXV which is involved in the ATP anchoring to the catalytic unit of all kinds of kinases. Subdomain IX contains an almost invariable Asp that acts to stabilize the catalytic curl, joining residues in the VIB subdomain. This invariant Asp flanking amino acids is frequently used in PCR cloning strategies, as it distinguishes βß-thymine-kinase (DVWSY / FGI / V) from serine / threonine-kinase (DXWA / SXGI / V). Based on the comparison of all known protein kinases, degenerate oligonucleotide sensitizers were designed for subdomains I and IX that collected only CCK4 and its chicken counterpart KLG, by PCR. We designed degenerate A and DVW sensitizers that are based on conserved residues within the kinase domain CCK4, for use in the identification of novel kinases, using the polymerase chain reaction (PCR). When applied to the HEPM cell sscDNA, as a template, multiple copies of CCK4 were isolated as well as a novel DNA fragment (43-43) of 567 base pairs, homologous with the other kinases. The novel sequence was very similar to Drosophila quinaa aurora (Accession No. GeneBank XB3465) and the human Auroral clone was designed. Using that fragment as a probe the RNAs of numerous colon cancer cell lines and a human multiple tissue Northern blot were discriminated, demonstrating an apparent selectivity in the expression of Auroral in tumor cells. The Auroral probe was also used to discriminate a cDNA library constructed from the mRNA of the human pancreatic cancer cell line to isolate the overlapping clones that cover the entire open reading frame of Auroral. Of the multiple isolated clones »seven correspond to human Auroral. Two additional weakly hybridizing clones were also isolated during this discrimination, and the sequence analysis revealed that they corresponded to a related "though distinct" kinase that was designated human Aurora2. AUR0RA1 and AUR0RA2 recombinants were expressed in COS cells migrated with apparent Mr of 39,000 and 46,000. consistent with their predicted molecular weights of 39264 and 46730. based on their primary amino acid sequence. This analysis confirms that the recombinant protein can be produced stably in mammalian cells. Phosphorylation assays to determine the target specificity of these putative kinases are being carried out. Specific immunoreactive were generated in rabbits against the peptide sequence of N-terminal domains of AUR1 and AUR2 »and these reagents were used to localize the expression of endogenous and recombinant Auroras within the cells. Additionally, that can be used reactive in an effort to identify the substrates for the AURORAS in order to better understand its normal biological role. The dominant and highly active negative form of AUR1 and AUR2 will be useful to delineate biological consequences in the oblation and over-expression of these putative serine / threonine-quinaßaß. Initial studies with altered DNA constructs demonstrate that in β aol 2 hours after infection of NIH3T3 or BALB / 3T3 cells with retroviral materials of AUR1 and AUR2, the cells remained highly tinucleated. This phenotype persisted in such a way that two days after infection it was found that some cells had up to 20 nuclei. The multifluted cells typically had increased cytoplasm and diffuse cell boundaries. Immunostaining with actin and with DAPI confirmed that these nuclei were all contained within a cell, and that the actin cytoskeleton was apparently normal. The experiments that are being carried out will direct the content of the chromosome and the intact within the nucleus, to the number and location of the centroßome and to the general organization of the icrotubular network. The characterization with long-term sequences of the expression of normal, constitutively active and negative dominant AUR1 and AUR2 is under study, and particularly if they induce or reverse cell transformation. It is isolating stable clones that express those recombinant proteins before. They will be characterized by the speed of growth, by the synthesis of DNA »the inhibition of contact with the cell (formation of foci)» the independent development of the anchor (analysis in soft agar) and the tumoricity in mice without hair. What role do Auroras play in the reproduction and segregation of mammalian centroßoma? The interruption or deregulation of these functions is known to result in the lack of aggregation of chromoßomaß »monopolar spindles and polar division in yeast» drosophila and amphibians. The homology between human Auroras and the yeast aurora IPL1 and drosophila is shocking. Therefore »it is convenient to determine if the human Auroras are functional equivalents of the yeast IPL1. The yeast IPL1 gene is necessary for chromosome segregation with high fidelity in 3Saccharomyces cerevisiae (Francisco, L. and co-authors, Mol Cell Biol. 14: 4731-4740 (1994)) and a temperature-sensitive mutant has been isolated . It is planned to determine if different full length and chimeric versions of the human Auroras can complement this mutant senεible to the temperature in yeast. Chimeric constructions containing the N-terminal domain of IPL1 and the kinase domain of human Auroraß will make it possible to determine ßi the kinase is functionally equivalent, while avoiding the regulatory roles or intracellular localization mediated by N-terminal domains. nales less well preserved. In case the human genes compensate for the loss of function IPL1, such a line could be used to discriminate the small molecule inhibitors of the human Aurora kinase.
MOLECULAR CLONING Total RNA was isolated using the guanid na / phenol salt extraction protocol from Chomczynski and Sacchi (P. Cho czynski and N. Sacchi, Anal. Biochem. 162, 156 (1987) from normal human prostate, duodenum, ovary, pituitary liver, brain, thymus and salivary glands, from human HEPM cells (palatal meßenchyma), from Wilm's primary human tumor and carcinoma of the ovaries, and from the human tumor cell lines originating from the colon / rectum (HT29, SW480, SW1463, SW1417, SW837, SW94B, SW620, SW403, SW111G. TB4 »HTC15» LS123 and caco-2) »kidney (CaKi-1, CaKi-2), liver (SK-HEP-1), páncreaß (HS766T, ASPC» Capan-1) and breast (MCF7) These RNAs were used as a template to generate the single-filament cDNAs using the superscript preamplification plan for the first-strand synthesis kit purchased from GibcoBRL (Life Technologies, USA, Gerard, GF and coautoreß, 1989). ), FOCUS 11, 66) under the conditions recommended by the manufacturer.A typical reaction used 10 μg total of RNA or 2 g poly (A) * RNA with 1.5 ug ol igo (DTT) i? _iß, in a reaction volume of 60 ul. The product was treated with aRNseH and εe diluted to 100 μl with Ha0. For the subsequent PCR amplification, 1 to 4 μl of eßtos sscDNA was used in each reaction. The o-nucleotides were synthesized in an Applied Biosystems 394 DNA synthesizer, using established phosphoramidite chemistry, and then used without purification after purification with ethane. The degenerate oligonucleotide sensitizers are: A = 5 »-GARTTYGGNGARGTNTTYYTNGC-3 '(SEQ ID NO: 16) (from 6B same sense) and DVW ß 5 »-AGNACNCCRAANGCCCACACRTC-3» (SEQ ID N0: 17) (opposite sense). These sensitizers were derived from the peptide sequences EFGEVFLA (SEQ ID NO: 18) (strand of the same direction from the kinase I subdomain) and DVW (A / S) FGVL (opposite sense strand from the kinase IX domain), respectively. The designations of degenerate nucleotide residue are: N = A »C. G or T; R = A or G; and Y = C or T. Using CCK4 as a template »these sensitizers produce a product of 567 base pairs. A PCR reaction was carried out using the Sensitizer 1 and DVW applied to the single-source source indicated above. The sensitizers at a final concentration of 5 μmol each were added to a mixture containing 10 mmole of Tris-HCl (pH 8.3). 50 mmole of KCl. 1.5 mmol of MgCl3 »200 μmol of each of deoxynucleoside triphosphate» 0.001% of gelatin and 1.5 units of AmpliTaq DNA-polymerase (Perkin-Elmer / Cetuß) and 1 to 4 μl of cDNA. After denaturation at 95 ° C for 3 minutes »the cycle conditions were 94 ° C for 30 seconds» 37 ° C for 1 minute »a ramp of 2 minutes up to 72 ° C and 72 ° C for 1 minute during 3 minutes first cycles »followed by 94 ° C for 30 seconds» 50 ° C for 1 minute and 72 ° C for 1 minute 45 seconds, for 35 cycles. The fragments of RCP that migrate to between 500 and 600 base pairs were affixed from 2% agarose gels using GeneClean (BiolOl) "and cloned by T-A in the vector pCRII (Invitrogen Corp. USA). according to the manufacturer's protocol. The colony was selected for miniplassed DNA preparations using Qiagen columns and the plasmid DNA sequence was determined using the dye terminator kit »cycle sequence deterrnant using AmpliTaq DNA-pol imerase» FS (ABI »Foster City» CA »USA). The reaction products of the sequence determination were run on the ABI Prism 377 DNA sequencer and analyzed using the BLAST alignment algorithm (Altschul »S.F. and co-authors» J. Mopl.Biol. 215: 403-10). A novel clone (843-43) was isolated by PCR with sensitizers A and DVW in single-stranded cDNA from the human embryonic palaennial mesenchyme (HEPM or CRL1486) as a template. This clone was designated as a fragment of human Auroral. A lambda ZapII cDNA bank was built (Stratagene Cloning Systems, La Jolla, CA, USA) using mRNA from a collection of pancreatic carcinoma cell lines as a template for the synthesis of first filament cDNA. The phage was discriminated in microcellulose filters with the insert marked with aP, randomly sensing 1, from p43-43 encoding human Auroral at 2x10 * cpam / l, in hybridization buffer containing βxSSC. l? Denhardt reagent, 0.1% SDS »with 0.1 mg / ml fragmented and denatured salmon epsperm DNA. After hybridizing overnight at 65 ° C »the filters were washed in O.lxSSC» 0.1% SDS at 65 ° C. The sequence of full length cDNA clones in both strands was determined, using manual sequence determination with T7 polymerase and oligonucleotide sensitizers (Tabor and Richardson, 1987, Proc. Nati. Acad. Sci. USA »84: 4767-71 ).
EXAMPLE 2 PE NORTHERN PE STAIN ANALYSIS Northern blotting containing 2 μg of poly A + RNA per lane was obtained from 16 different adult human tissues (spleen »thymus» prostate »testicle» ovary »small intestine» colonic mucosa »heart» brain »placenta» lung » liver »skeletal muscle» kidney »pancreas and peripheral blood leukocytes), four different human fetal tissues (brain, lung, liver and kidney) and 8 human cancer cell lines (HL60, HeLa, K-562, MOLT-4. Raji, SW480 and G361) in a nylon membrane modified with charge, from Clontech (Palo Alto, CA. USA). Additional Northern blots were prepared by operating 10 μg of total RNA isolated from human tumor cell lines, in a 1.2% agarose gel with formaldehyde. denaturing "and transferring to nylon membranes. The filters were hybridized with random preparation probes, marked with C ^^ PDdCTP. synthesized from either the 427 base pair insert of human Auroral clone 43-43 or the 1162 base pair EcoRI fragment from PSG20 and 1 kb EcoRI fragment from human Aurora2 clone 11-1A or the Ba HI- fragment Not I of 1257 base pairs, of PS621. Hybridization was carried out at 60 ° C in 6XSSC. 0.1% of SDS »IX of Denhart's solution. 100 mg / ml of herring sperm DNA denaturally hoisted with 1-2? 10 * cpm / ml of DNA probes labeled with 3aP. The filters were washed in 0.1XSCC / 0.1% SDS »65 ° C» and exposed over night on Kodak XAR-2 film. A single transcript of mRNA AUR1 »of approximately 1.4 kb was identified and found to be more abundant in the thymus and in the small intestine» with weak testicular »ovarian» colon »placenta and spleen. The prostate and the peripheral blood lymphocytes were negative. The human fetal liver and kidney were also positive with the weakest signal in the fetal lung and no expression in the fetal brain (table). A similar analysis of the expression of human AUR2 or a more restricted e? Preion profile. A translocated ßlolo of AUR2 »2.4 kb» was detected strongly in the adult testis and thymus »and weakly in the cardiac cluster» of placenta and skeletal D, and in the fetal liver and in the kidney »while in other tissue sources normal were negative (see box).
ANALYSIS OF NORTHERN OF AURORA 1 AND AURORA 2 IN NORMAL TEUIPO CELLS AND HUMAN CANCER CELLS Type of cells Origin AUR1 AUR2 Thymus Normal tissue 5 4 Fetal liver Normal tissue 4 2 Fetal kidney Normal tissue 4 1 ' Lung Tissue norma1 3 0 Duodenum Normal tissue 2 1 Colon Normal tissue 2 0 Fetal lung Normal tissue 2 0 Ovaries Normal tissue 2 0 Testicles Normal tissue 2 2 Brain Normal tissue 0 0 Cerebellum Normal tissue 0 0 Salivary gland Normal tissue 0 0 Heart Normal tissue 0 0 Liver Normal tissue 0 0 Pancreas Normal tissue 0 0 Kidney Normal tissue 0 0 Spleen Normal tissue 0 0 Stomach Normal tissue 0 0 Uterus Normal tissue 0 0 Prostate Normal tissue 0 0 Skeletal muscle Normal tissue 0 0 Fetal brain Normal tissue 0 0 Type of cells Origin AUR1 AUR2 PBL Normal fabric 0 0 Salivary gland Normal tissue 0 0 Placenta Normal tissue 0 0 SF-268 CNS tumor 4 ND CCRF-CEM Leukemia 4 ND K-5G2 Leukemia 4 ND HCC-2998 Colon Tumor 4 ND SW620 Colon Tumor 4 2 KM-12 Leuce ia 4 ND MCF7 / ADR-RES Breast tumor 4 2 MDA-N Breast tumor 4 ND BT-549 Breast tumor 4 ND SW4B0 Colon tumor 4 4 SW48 Colon tumor 4 ND CalU-3 Pulmonary Tumor 4 ND Calu3 Lung tumor 4 2 T47D Breast tumor 4 2 A375 Melanoma 4 0 SF767 Tumor of 1 SNC 4 0 SW1417 Colon tumor 4 4 CaKi2 Kidney tumor 4 0 Ca il Kidney tumor 4 0 Caco2 Colon tumor 4 4 SW1417 Colon tumor 4 0 T98G Tumor of 1 SNC 4 0 Type of cells Origin AUR1 AUR2 SF-539 Tumor of 1 SNC 3 ND SK-MEL-2 Melanoma 3 ND SK-MEL-5 Melanoma 3 ND R-4B Gastric tumor 3 ND RF-1 Gastric tumor 3 ND SW948 Colon Tumor 3 ND AGS Gastric tumor 3 ND HFL1 Normal lung 3 ND OVCAR-8 Ovarian tumor 2 ND HT-29 Colon Tumor 2 ND MDA-MB-231 Breast tumor 2 ND MDA-MB-435 Breast tumor 2 ND SK-MEL-5 Melanoma 2 ND Kato-3 Gastric tumor 2 ND ColO 205 Colon Tumor 2 ND ColO 320DM Colon tumor 2 2 WiDr Colon Tumor 2 ND HT-29 Colon Tumor 2 ND SNU-C2B Colon Tumor 2 ND HTC15 Colon tumor 2 2 T84 Colon tumor 2 0 SW948 Colon tumor 2 0 Daoy Tumor dell CNS 2 0 0VCAR3 Ovarian tumor 2 0 HS766T Pancreatic tumor 2 0 Type of cells Origin AUR1 AUR2 SW1116 Colon tumor 2 0 Wilm's Tumor Kidney Tumor 2 0 UO-31 Kidney Tumor 0 ND The e1-preεion profile of AUR1 mRNA was determined in varioß primary tumors and in several multiple cell lines »of various neoplastic origin» by Northern analysis and by Semi-quantitative PCR analysis »using sensitizers from the sequences in the kinase domain of AURl. The results are included in the Table. The AUR1 transcripts were detected in each tumor line analyzed with the highest expression in several human cancer cell lines (SW480, Colo320 »SW620» SW147 »Caco2» SW12417) and in lung carcinoma (Calu3) > Mema carcinoma (T47D, MCF7). melanoma (A375). kidney carcinoma (CaKi-1, CaKi-2) »liver carcinoma (SK-HEP-1) and in neuraleß tumors (SF767» T9BG). The lower expression of AUR1 was observed in other colon carcinomas (HTC15, TB4, SW94T, SW1116, HT29). Neural tumors (Daoy). Ovarian carcinoma (0vcar2, primary tumor), pancreatic carcinoma (HS766T) and a primary kidney tumor. The expression profile of AUR2 in the tumor cell lines was shockingly more restricted than that of AUR1. Strong expression of AUR2 was detected only in the lines of colon carcinoma cells (Caco2, SW1417 »SW620) while weak lines were observed in other colon (HTC15» Colo320) »breast (T47D, MCF7 ) and lung (Calu3). Other different tumor lines did not have detectable AUR2 transcripts.
EXAMPLE 3 PET PETION RCP SEMICUANTITATIYA PE AURORA! RNA was isolated from a variety of human cell lines - fresh frozen tissues and primary tumors. The single-stranded cDNA was synthesized from 10 mg of each RNA, as described above, using the Superscript Preemplification system (GibcoBRL). These single-filament templates were then used in a 35-cycle PCR reaction, with two specific oligonucleotides of AUR0RA1 (3476: 5 »-TTTGGCTCGGGAGAAGAAAAGCCAT-3» (SEQ ID N0: 19) and 3506: 5'-CATCATCTCTGGGGGCAGGTAGT- 3 ') (SEQ ID N0: 20) The reaction products were electrophoresed on 2% agarose gels, stained with ethidium bromide and photographed in a UV light box. Specific bands of Auroral of approximately 475 base pairs »for each sample.
EXAMPLE - SOUTHERN SPOT ANALYSIS Genomic DNA was isolated from a variety of trans human human lines (CaC02 * HTC15, LS147T, SKC04, SW480 »SW403, SW620, SW948» SW1417 »SW1116» MCF7 »BT474) using common procedures (Maniatis and coauthors). Cells were triptinized, washed with PBS and resuspended to about 10 cells / ml in digestion buffer (100 mmoles of NaCl, 10 mmoles of Tris pH 8, 25 mmoles of EDTA, pH 8, 0.5%). SDS »0.1 mg / ml proteinase K) The cells were lysed by incubation at 50 ° C for 12 hours, followed by extraction with phenol / chloroform and precipitated with an equal volume of 7.5 moles of ammonium acetate and 100 % EtOH The DNA was re-suspended in the TE buffer Approximately 20 micrograms of genomic DNA was digested with HindIII or XhoII at 37 ° C for at least 4 hours before fractionating on 1% agarose gels. fragments of DNA to membranes of no troceuyosa, by means of the capital transfer method (Southern, EM, J. Mol, Bio, 98: 503, 1975) and hybridized with specific probes for Auroral and human Aurora2, as described above. for the analysis of previous Northern blot, DNA was restricted with HindIII since the cDNAs of AUR1 and AUR2 contain a single site for this restriction enzyme. AUR1 showed a single band of 4.3 kb of equal intensity »from all sources» suggesting that it is a single non-rearranged gene »of a single copy» in the multiple tumor types analyzed. Nevertheless, under conditions of low demand »it was possible to detect Sacl fragments of 1.3 kb and 3.2 kb» that hybridized weakly to the AURI probe. Cloning and sequence analysis revealed that this region encoded a pseudogene related to AUR1 »without intron (termed AUR3)» with multiple frame shifts. Additionally »immediately above the AUR3 pseudogene» there is a region with complex inverted repeats »which was predicted to form a very stable hairpin curl. The DNA sequence of AUR3 eß homologates with AUR1 »starting from the first nucleotide of the cDNA of AUR1. Immediately upstream of this site »a hairpin loop of AUR3 is predicted. The genomic clones of AUR1 are currently being characterized to determine whether the homology with AUR3 continues upstream of this nucleotide and whether the DNA of AUR1 includes or is preceded by a similar hairpin curl. AUR2 showed bands at 7.0 kb and 4.3 kb and a weak band of higher molecular weight at around 10 kb, from all sources. These data suggest that AUR2 eß also a gene of a beta copy. The multiple bands that were seen in the blots probed with AUR2 are probably due to the fact that a full-length cDNA probe was used.
EXAMPLE 5 ANALYSIS OF SEQUENCE OF CADN CLONES THAT CODE HUMAN AURORAl AND AURQRAZ The complete sequence of human Auroral and Aurora2 was determined from full-length clones of each of the isolates of the human pancreatic carcinoma »bank from normal human duodenum» and of partial human Auroral isolated from HEPM cells. The nucleotide sequence of human Auroral (AURl_h) of 1,244 base pairs »is shown in SEQ ID N: i or SEQ ID N0: 2 and contains a single" open reading frame "that encodes a 344 amino acid polypeptide. The coding region of AURl_h is flanked by a 5 * untranslated region of 54 nucleotides and a 3 'untranslated region of 132 nucleotides »ending with a tail of pol i (A). The nucleotide sequence of human Aurora2 (AUR2_h) > of 2 »198 base pairs» is shown in SEQ ID NO: i or SEQ ID NO: 2 and contains a single open reading frame, which encodes a 403 amino acid polypeptide. The coding region of AUR2_h is flanked by a non-transfected 5T region of 200 nucleotide and a non-transcribed region of 768 nucleotides. The cDNA sequences of AUR1 and AUR2 were determined from human pancreatic tumor and normal duodenum, without sequence differences except for some sites, probably pol imorphicß. Eßta ambiguities include: cDNA Nucleotide Comment or AUR1 1174 one clone has poly A 873 T insert in all duodenal clones »C in pancreatic tumor 469 T in one clone» C in all other 848 G in one clone »A in all others.- amino acid changes E a G 1097 G in one clone »T in another 2 956 G in one clone» A in others 4 29 Splicing in 103 in 5 clones »no splicing (as shown in 5 clones) AUR2 349 T in 1 clone. C in other multiple (the amino acid changes P to L) 369 A in 3 clones »G in other multiple (changes AA V to I). The C terminal portions of AUR1 and AUR2 retain all 12 subdomains characteristic of eukaryotic protein kinases. These kinase domains of AUR1 and AUR2 are preceded by an N-terminal domain of 74 and 130 amino acids, respectively. nucleotide AUR1 was compared and AUR2 and the amino acid sequences were deduced (SEQ ID NO: 3 or SEQ ID N0: 4) with the available DNA and the base datasets of protein sequences that are unique »with the exception of several EST sequence» that share a high sequence identity. However, they do have a shocking homology in both the N. terminal and catalytic domains with drosophila aurora and with the IPL1 genes of Saccharomyces cerevisiae. Additional »two annotations from the unpublished database are likely to be close homologues of Xenopus laevis (p46APK - access number of GB Z1726 and p46BPK - accession number of GB 217207). The N-terminal domains of Aurors derived from human »frog» from Drosophila and from yeast share a limited sequence identity. AUR2 of requirement has an abundance of glutamates frequently preferentially in paree separated by a single residue. The comparison of the catalytic domains of these proteins reveals that AUR1 shares 70% identity of amino acids with AUR2 »61% with Drosophist Aurora and 45% with the yeast IPL1 gene. The AUR2 kinase shares 60% amino acid identity with the Drosophila protein and 45% identity with yeast IPL1. Both AUR1 and AUR2 share less than 45% homology with all other known mammalian kinases (the cAMP-dependent protein kinase A eß the closest) "suggesting that they are homologues of these Drosophila kinase and yeast. AUR1 and AUR2 both contain a protein kinase phosphorylation site, dependent on cAMP (THR232 of AUR1 and THR288 of AUR2). which is conserved in the drosophila and levßdura homologs and a regulatory site known in the cyclin-dependent kinase p34cdc2. AUR2 contains an additional PKA site in SER342. Both proteins also have multiple phosphorylation sites of casein kinase II (five and six for AUR1 and AUR2) and protein kinase C (four and ten for AUR1 and AUR2). AUR2 also has a single tyrosine phosphorylation consensus site in TYR334 that also retains drosophila aurora. but it is not present in AUR1 or in yeast IPL1. In an intriguing way, the natural mutants of the drosophila dawn AUR_dm and the yeast ILP1 gene result in the asymmetric nuclear division leading to erroneous chromosome segregation and to atypical monopolar spindles. This phenotype seems to be the result of a failure in the separation of the centrosome. The associated icrotubular architecture seems to be unaffected. The natural mutants in both the drosophilic and the yeast destination amino acid residues, which are strictly conserved among the human Auroras, further support that they are functional homologues. The corresponding residues in AUR1 »found in natural mutant eß of AUR_dm or IPL1» ßon GLU125 »THR232» PR0312 »HIS324. All these mutations are within the catalytic domain and »primarily» one represents the conserved PKA phosphorylation site. An additional mutation in AUR_dm in ASP47 »is in a non-conserved residue in the N-terminal domain. These findings suggest that catalytic activity may actually play a central role in the biology of centrosomal reproduction or segregation in lower organisms, and suggests that human auroras may play a complementary role in mammalian cells.
EXAMPLE ß RECQMBINANT EXPRESSION PE CONSTRUCTION PEL VECTOR PE EXPRESSION PE AURORA! AND AURQRAZ Expression constructs were generated by PCR-assisted mutagenesis in which the entire coding domains of Auroral and Aurora2 were labeled at their carboxy-terminal ends with the YPYDVPDYAS epitope (SEQ ID N0: 21) (Pati »1992) of the hemagglutinin (HA) of hemophilic influenza. These constructs were introduced into two mammalian expression vectors: pLXSN (Miller, A.D. and Roßman, G.J. Biotechniques 7, 980-988, 1989) for the generation of virus-producing lines; and pRK5 for the analysis of transient expression. Loe inßertoß was designed to be flanked by the unique BamHI and Notl sites. and were cloned directly to pLXSN or pRK5 »in the ßitioß 5 * -BamHI and 3'-NotI. The full-length AUR1 and AUR2 constructs were also ligated from BamHl-Notl to pRS316 (Liu »H. and coauthors» Genetics 132: 665-673 »1992). This vector contains a galactose-inducible promoter in a centromeric back-and-forth vector for expression in Saccharomyces cerevisiae. It remains to be determined whether human genes can complement the related »temperature sensitive» yeast IPL1 mutant that is intimately related to AURl. Further »fusion constructs containing the N-terminal domain of yeast IPL1» fused to »the C-terminal kinase domains of AUR1 and AUR2» were generated. These were produced by insertion of an artificial Clal ßitium at the 5 'end of the kinase domains of the kinases. in the conserved Aßp-Asp-Phe-Glu sequence. The "dominant" negative AUR1 and AUR2 were also made in both pLXSN and pRK5 > by invariant Lys mutation (amino acid positions 106 and 162 in AUR1 and AUR2, respectively) »to a Met by PCR mutagenesis. The constructions were called AUR1KM and AUR2KM. Constitutively active forms of AUR1 and AUR2 were generated by mutation of the DNA that leads the encoding of the phosphorylation site (232 and 288). to an Aßp that results in AUR1TD and AUR2TD. Expression constructs were also made in both pLXSN and pRK5 that contained only the N-terminal non-catalytic domain of AUR1 and AUR2. Eßtoß were generated by PCR from environmental constructions and containing the 77 N-terminal amino acids of AUR1 and the 132 amino acids of AUR2. The complete open reading frames of AUR1 and AUR2 (without HA tag), excluding initiator methionine, were generated by PCR and bound to the pGEX vector for the bacterial production of GST fusion proteins, for the immunization of rabbits, for the production of antibodies.
EXAMPLE 7 SENERATION PE LINES PE PE CELLS AURORA PRQPUCTQRAS PE VIRUS To generate high-titre viruß materials, ß transfected recombinant pLXSN constructs containing AUR1 or AUR2 genes. to a line of PA317 »amphotropic helper cells using CaCl-mediated transfection. After selection in G418» plaques were formed with the cells in normal media G418 (500 μg / ml). The supernatants of the reßißtentß cells were used to infect the GP + E86 ecotropic helper cell line and again selected the cells in G41B. The reßißtenteß cells of G418 were harvested again and the supernatants harvested every 8 to 12 hours and were pooled as virus material (Rede ann, N., Holzmann, B., Wagner, EF, Schlesinger, J. and Ullrich , A., Mol. Cell, Biol. 12, 491-498, 1992). Titers of viral material were typically around 10 * / ml.
EXAMPLE fl RETROVIRAL INFECTION OF NIH-3T3 CELLS WITH AURORAS NIH-3T3 and BALB / 3T3 cells were developed in 100-mm dishes with DMEM (Gibco) containing 10% fetal calf serum (FCS). The cells were superinfected with retroviruses of AUR1 and AUR2, adding approximately 3 ml of the viral supernatant to 15 ml culture media, for approximately 24 hours. The cells expressing the retroviral constructs were then selected to be developed in DMDM / 10% FCS, supplemented with 500 μg / ml of G418.
EXAMPLE 9 GENERATION OF AURORA SPECIFIC IMMUNOREABLE Immunoreactive specifics were raised for AURORA in rabbits, against synthetic peptides conjugated in KLH »corresponding to the N-terminal region of AUR2 (xo- ^ SAPENPEEQLASK ** - *) (SEQ ID NO: 22) and (• ° RPLNNTQKSKQPLa- ° *) (SEQ ID N0: 23). or the N-terminus or the N-terminal domain of human AUR1 (^ MAQKENSYPWPYG3- ») (SEQ ID NO: 24) and (" PGWKVMENSSGTP * ") (SEQ ID NO: 25). Additionally, immunoreactives were generated by immunizing rabbits with the fusion proteins of AUR1 and AUR2 with full-length GDT expressed bacterially.
EXAMPLE 10 TRANSIENT EXPRESSION PE AURORAS IN MAMMAL PE CELLS The expression plasmids pRK5 (lOμg DNA / 100 mm plate) containing the HA-tagged AUR1 and AUR2 genes were introduced. in COS cells and 293. with lipofectamine (Gibco BRL). After 72 hours the cells were harvested in 0.5 ml of solubilization buffer (20 mmoles of HEPES pH 7.35 »150 mmoles of NaCl, 10% glycerol, 1% Triton X-100, 1.5 mmoles MgCl-, 1 mmole. of EGTA »2 mmoles of feni lmethylsulfonyl fluoride» 1 μg / ml of aprotinin). The sample counts were solved by SDS »electro polyacrylamide gel electrophoresis (PAGE) on 15% acri lamide / 0.5% bis-acri lamide gels» and electrophoretically transferred to nitrocellulose. The non-specific binding was blocked by pre-spotting the Blotto stains (phosphate-buffered saline containing 5% w / v dry milk defatted and 0.2% volume / volume of nonidet P-40 (Sigma)) »and detected the recombinant protein using a murine Mb to the decapeptide label of HA. Alternatively, it can detect the recombinant protein using various antisera specific for AUR1 or AUR2.
EXAMPLE 11 BASIC MYELINE PROTEIN IS AN ARTIFICIAL SUBSTRATE FOR LA QUI ASA DE AURORAl AND AUR0RA2 Method SW480 cells from human colorectal adenocarcinoma were cultured in RPMI 1640 plus 10% fetal bovine serum. L- 8B glutamine, penicillin and streptomycin. Confluent cultures of SW480 cells were washed three times with ice-cold phosphate buffered saline (PBS), and then scraped into 1 ml of ice-cold PBS. The cells were centrifuged at 1000 rpm at 4 ° C and the PBS was aspirated, and the resulting cell pellet was stored at -80 ° C. The pellets of three 15 cm plates were thawed on ice and resuspended in a total of 1 ml of kinase lisie regulator »50 mmoles of HEPES» pH 7.4 »100 ml of KCl» 25 mmoles of NaF »1 mmol of NaV0a »0.5% of NP40» 1 mmol of DTT »2 μg / ml of aprotinin and 1 μg / ml of leupeptin)» and was rotated gently for 20 minutes at 4 ° C. The samples were then centrifuged at 10 000 x g for 10 minutes at 4 ° C and the resulting supernatant was transferred to a clean centrifuge tube of 1.5 m and stored and kept on ice. Protein concentration was determined by Bradford analysis. 1 mg of total protein was pre-cleared with 10 μl of protein A-Sepharose (Boehringer) for 15 minutes at 4 ° C followed by 2 μl of rabbit preimmunized serum »affinity purified auroral peptide antiserum, purified auroral peptide antiserum by affinity plus 6 μg of competing auroral peptide, affinity purified aurora2 antiserum or affinity purified aurora 2 peptide antiserum plus 6 μg of competitor aurora 2 peptide, and incubated for 30 minutes at 4 ° C. Subsequently, 10 μl of protein A-Sepharose was added and incubation was continued for another 45 minutes at 4 ° C. The tubes were briefly centrifuged to form pellets with the antibody-protein A-Sepharoße complex and the resulting supernatant was aspirated. The antibody-protein A-Sepharose pellet was washed twice with 0.5 ml of kinase lysis buffer »followed by a wash with 0.5 ml of kinase regulator (20 mmoles of HEPES pH 7.4» 125 mmoleß of KCl »10 mmoles of MgCla, 1 mmol of NaF »1 mmol of NaV0a and 1 mmol of DTT). The antibody-protein A-Shefaróse pellet was resuspended in 20 μl of kinase regulator containing 5 μCi of Cgamma-3aP3 ATP and 0.5 mg / ml of myelin basic protein (Sigma) was incubated for 20 minutes at 37 ° C »after which 10 μl of protein buffer was added» 200 mmoleß of Tris-HCl pH 6.8, 40% glycerol, 730 mmol B-mercaptoethanol, 0.4% SDS and 0.05% bromophenol blue). The tubes were mixed well and incubated for 5 minutes at 100 ° C. The samples were resolved in an 18% SDS-polyacrylamide gel and visualized by autoradiography.
LQft resu ats The immunocomplexes of auroral and aurora2 were able to phosphorylate the myelin basic protein. When a competing peptide was used in the immunoprecipitations, none of the auroral or aurora2 anti-serum immunocomplexes were able to phosphorylate the myelin basic protein more than the control of pre-immunological serum. This suggests that the kinase activity observed is due to auroral and 2, and not to other proteins present in the intraununocomplex. This observation will allow the purification of auroral kinase and 2 using the myelin basic protein as a substrate to follow the kinase activity. It will also allow the development of an in vitro kinase analysis using auroral and 2 recombinant proteins. Additionally, an analysis of auroral kinase and 2 in vitro will allow to discriminate the collections of small molecules for inhibitors of auroral kinases and 2. to measure the inhibition of phosphorylation in the basic protein of honey ina. The invention illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations that are not specifically described herein. The terms and expressions that have been used are terms of description and not of limitation, and it is not intended that in the use of said terms and expressions, any equivalent of the aspects shown and described or portions thereof will be excluded. »But it is recognized that various modifications are possible within the scope of the claimed invention. Thus »it should be understood that» although the present invention has been described specifically by the preferred modalities and optional aspects, it is possible to resort to the modification and variation of the concepts described herein by the person skilled in the art, and that said modifications and variations are considered to be within the scope of this invention, as defined in the claims that follow. References not previously incorporated herein by reference "including both patents and non-patent references" are expressly incorporated herein by this reference "for all purposes. Other modalities are within the claims that come at the end.
LISTAPO PE SEQUENCES (1) GENERAL INFORMATION: (i) APPLICANT: Mossie »Kevin G. Plowman, Gregory D. (ii) TITLE OF THE INVENTION: DIAGNOSIS AND TREATMENT OF ALTERATIONS RELATED TO AUR-l AND / OR AUR-2 (iii) NUMBER OF SEQUENCES: 6 (iv) ADDRESS FOR CORRESPONDENCE: (A) NAME: Lyon & Lyon (B) STREET: 633 West Fifth Street »Suite 4700 (C) CITY: Los Angeles (D) STATE: California (E) COUNTRY: USA (F) ZIP CODE: 90071-2066 (iV) FORM (v) ) WHAT CAN BE READ IN COMPUTER: (A) TYPE OF MEDIUM: flexible disk (B) COMPUTER: IBM compatible PC (C) OPERATING SYSTEM: PC-DOS / MS-DOS (D) APPLICATION PROGRAM: Patentln Relay 1 * 1.0 »Version tl.30 (V) FACTS ABOUT THIS APPLICATION: (A) APPLICATION NUMBER: US (B) SUBMISSION DATE: (C) CLASSIFICATION: (Vii i) EMPLOYEE / AGENT INFORMATION (A) NAME: Wargurg. Richard J. (B) REGISTRATION NUMBER: 32 »327 (ix) TELECOMMUNICATIONS INFORMATION: (A) TELEPHONE: (213) 489-1600 (B) TELEFAX: (213) 955-0440 (C) TELEX: 67-3510 (2) INFORMATION FOR SEQ ID N?: L (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 1244 base pairs (B) TYPE: nucleic acid (C) NUMBER OF FILAMENTS: one ONLY (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: CADN (iii) HYPOTHETICAL: NO (iv) CONTRARY SENSE: NO (vi) NATURAL SOURCE: (A) ORGANISM: Homo sap ens (Xi) DESCRIPTION OF THE SEQUENCE: SEQ ID N0: i CGGGAGAGTA GCAGTGCCTT GGACCCCAGC TCTCCTCCCC CTTTCTCTCT AAGGATGGCC 60 CAGAAGGAGA ACTCCTACCC CTGGCCCTAC GGCCGACAGA CGGCTCCATC TGGCCTGAGC120 ACCCTGCCCC AGCGAGTCCT CCGGAAAGAG CCTGTCACCC CATCTGCACT TGTCCTCATG180 AGCCGCTCCA ATGTCCAGCC CACAGCTGCC CCTGGCCAGA AGGTGATGGA GAATAGCAGT2 0 GGGACACCCG ACATCTTAAC GCGGCACTTC ACAATTGATG ACTTTGAGAT TGGGCGTCCT300 CTGGGCAAAG GCAAGTTTGG AAACGTGTAC TTGGCTCGGG AGAAGAAAAG CCATTTCATC360 GTGGCGCTCA AGGTCCTCTT CAAGTCCCAG ATAGAGAAGG AGGGCGTGGA GCATCAGCTG420 CGCAGAGAGA TCGAAATCCA GGCCCACCTG CACCATCCCA ACATCCTGCG TCTCTACAAC 80 TATTTTTATG ACCGGAl-3AG GATCTACTTG ATTCTAGAGT ATGCCCCCCG CGGGGAGCTC 540 TACAAGGAGC TGCAGAAGAG CTGCACATTT GACGAGCAGC GAACAGCCAC GATCATGGAG600 GAGTTGGCAG ATGCTCTAAT GTACTGCCAT GGGAAGAAGG TGATTCACAG AGACATAAAG660 CCAGAAAATC TGCTCTTAGG GCTCAAGGGA GAGCTGAAGA TTGCTGACTT CGGCTGGTCT720 GTGCATGCGC CCTCCCTGAG GAGGAAGACA ATGTGTGGCA CCCTGGACTA CCTGCCCCCA780 GAGATGATTG AGGGGCGCAT GCACAATGAG AAGGTGGATC TGTGGTGCAT TGGAGTGCTT840 TGCTATGAGC TGCTGGTGGG GAACCCACCC TTCGAGAGTG CATCACACAA CGAGACCTAT900 CGCCGCATCG TCAAGGTGGA CCTAAAGTTC CCCGCTTCTG TGCCCACGGG AGCCCAGGAC960 CTCATCTCCA AACTGCTCAG GCATAACCCC TCGGAACGGC TGCCCCTGGC CCAGGTCTCJk.020 GCCCACCCTT GGGTCCGGGC CAACTCTCGG AGGGTGCTGC CTCCCTCTGC CCTTCAATCT080 GTCGCCTGAT GGTCCCTGTC ATTCACTCGG GTGCGTGTGT TTGTATGTCT GTGTATGTAT140 AGGGGAAAGA AGGGATCCCT AACTGTTCCC TTATCTGTTT TCTACCTCCT CCTTTGTTTA200 ATAAAGGCTG AAGCTTTTTG TAAAAAAACA AAAAAAAAAA AAAA 1244 (2) INFORMATION FOR SEQ ID NO: 2 (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 2198 base pairs (B) TYPE: nucleic acid (C) NUMBER OF FILAMENTS: one SOlO (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: cDNA (iii) HYPOTHETICAL: NO (iv) CONTRARY SENSE: NO (Xi) DESCRIPTION OF SEQUENCE: SEQ ID N0: 2 GGGATATCTC AGTGGCGGAC GAGGACGGCG GGGACAAGGG GCGGCTGGTC GGAGTGGCGG 60 ACGTCAAGTC CCCTGTCGGT TCCTCCGTCC CTGAGTGTCC TTGGCGCTGC CTTGTGCCCG120 CCCU3CGCCT TTGCATCCGC TCCTGGGCAC CGAGGCGCCC TGTAGGATAC TGCTTGTTAC180 TTA TACAGC TAGAGGCATC ATGGACCGAT CTAAAGAAAA CTGCATTTCA GGACCTGTTA240 AGGCTACAGC TCCAGTTGGA GGTCCAAAAC GTGTTCTCGT GACTCAGCAA TTTCCTTGTC300 AGAATCCATT ACCTGTAAAT AGTGGCCAGG CTCAGCGGGT CTTGTGTCCT TCAAATTCTT360 CCCAGCGCGT TCCTTTGCAA GCACAAAAGC TTGTCTCCAG TCACAAGCCG GTTCAGAATC 20 AGAAGCAGAA GCAATTGCAG GCAACCAGTG TACCTCATCC TGTCTCCAGG CCACTGAATA480 ACACCCAAAA GAGCAAGCAG CCCCTGCCAT CGGCACCTGA AAATAATCCT GAGGAGGAAC540 TGGCATCAAA ACAGAAAAAT GAAGAATCAA AAAAGAGGCA GTGGGCTTTG GAAGACTTTG600 AAATTGGTCG CCCTCTGGGT AAAGGAAAGT TTGGTAATGT TTATTTGGCA AGAGAAAAGC660 AAAGCAAGTT TATTCTGGCT CTTAAAGTGT TATTTAAAGC TCAGCTGGAG AAAGCCGGAG720 TGGAGCATCA GCTCAGAAGA GAAGTAGAAA TACAGTCCCA CCTTCGGCAT CCTAATATTC780 TTAGACTGTA TGGTTATTTC CATGATGCTA CCAGAGTCTA CCTAATTCTG GAATATGCAC840 CACTTGGAAC AGTTTATAGA GAACTTCAGA A ACTTTCAAA GTTTGATGAG CAGAGAACTG900 CTACTTATAT AACAGAATTG GCAAATGCCC TGTCTTACTG TCATTCGAAG AGAGTTATTC960 ATAGAGACAT TAAGCCAGAG AACTTACTTC TTGGATCAGC TGGAGAGCTT AAAATTGCAO; 020 ATTTTGGGTG GTCAGTACAT GCTCCATCTT CCAGGAGGAC CACTCTCTGT GGCACCCTGS080 ACTACCTGCC CCCTGAAATG ATTGAAGGTC GGATGCATGA TGAGAAGGTG GATCTCTGGA140 GCCTTGGAGT TCTTTGCTAT GAATTTTTAG TTGGGAAGCC TCCTTTTGAG GCAAACACAT200 ACCAAGAGAC CTACAAAAGA ATATCACGGG TTGAATTCAC ATTCCCTGAC TTTGTAACAG260 AGGGAGCCAG GGACCTCATT TCAAGACTGT TGAAGCATAA TCCCAGCCAG AGGCCAATGC320 TCAGAGAAGT ACTTGAACAC CCCTGGATCA CAGCAAATTC ATCAAAACCA TCAAATTGCCC380 AAAACAAAGA ATCAGCTAGC AAACAGTCTT AGGAATCGTG CAGGGGGAGA AATCCTTGAG440 CCAGGGCTGC CATATAACCT GACAGGAACA TGCTACTGAA GTTTATTTTA CCATTGACTS500 CTGCCCTCAA TCTAGAACGC TACACAAGAA ATATTTGTTT TACTCAGCAG GTGTGCCTTA560 ACCTCCCTAT TCAGAAAGCT CCACATCAAT AAACATGACA CTCTGAAGTG AAAGTAGCCA620 CGAGAATTGT GCTACTTATA CTGGTTCATA ATCTGGAGGC AAGGTTCGAC TGCAGCCGCÍH680 CCGTCAGCCT GTGCTAGGCA TGGTGTCTTC ACAGGAGGCA AATCCAGAGC CTGGCTGT G < B740 GGAAAGTGAC CACTCTGCCC TGACCCCGAT CAGTTAA 'GA GCTGTGCAAT AACCTTCCTA800 GTACCTGAGT GAGTGTGTAA CTTATTGGGT TGGCGAAGCC TGGTAAAGCT GTTGGAATGA86O GTATGTGATT CTTTTTAAGT ATGAAAATAA AGATATATGT ACAGACTTGT ATTTTTTCT (R920 TGGTGGCATT CCTTTAGGAA TGCTGTGTGT CTGTCCGGCA CCCCGGTAGG CCTGATTGGfli980 TTTCTAGTCC TCCTTAACCA CTTATCTCCC ATATGAGAGT GTGAAAAATA GGAACACGTG0 0 CTCTACCTCC ATTTAGGGAT TTGCTTGGGA TACAGAAGAG GCCATGTGTC TCAGAGCTG2100 TAAGGGCTTA TTTTTTTAAA ACATTGGAGT CATAGCATGT GTGTAAACTT TAAATATGC & 160 AATAAATAAG TATCTATGTC AAAAAAAAAA AAAAAAAA 2198 (2) INFORMATION FOR SEQ ID NO: 3 (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 344 amino acids (B) TYPE: amino acids (C) NUMBER OF FILAMENTS: one ßolo (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: protein (iii) HYPOTHETICAL: NO (iv) CONTRARY SENSE: NO (xi) DESCRIPTION OF THE SEQUENCE: SEQ ID NO: 3 Met Wing Gln Lys Glu Asn Ser Tyr Pro Trp Pro Tyr Gly Arg Gln Thr 1 5 10 15 Wing Pro Be Gly Leu Be Thr Leu Pro Gln Arg Val Leu Arg Lys Glu 20 25 30 Pro Val Thr Pro Be Wing Leu Val Leu Met Ser Arg Ser Asn Val Gln 35 40 45 Pro Thr Wing Wing Pro Gly Gln Lys Val Met Glu Asn Being Ser Gly Thr 50 55 60 Pro Asp lie Leu Thr Arg His Phe Thr lie Asp Asp Phe Glu lie Gly 65 70 75 80 Arg Pro Leu Gly Lys Gly Lys Phe Gly Asn Val Tyr Leu Ala Arg Glu 85 90 95 Lys Lys Ser His Phe lie Val Wing Leu Lys Val Leu The Lys Ser Gln 100 105 110 lie Glu Lys Glu Gly Val Glu His Gln Leu Arg Arg Glu lie Glu lie 115 120 125 Gln Ala His Leu His Pro Asn lie Leu Arg Leu Tyr Asn Tyr Phe 130 135 140 Tyr Asp Arg Arg Arg lie Tyr Leu lie Leu Glu Tyr Ala Pro Arg Gly 145 150 155 160 Glu Leu Tyr Lys Glu Leu Gln Lys Ser Cys Thr Phe Asp Glu Gln Arg 165 170 175 Thr Ala Thr lie Met Glu Glu Leu Wing Asp Wing Leu Met Tyr Cys His 180 185 190 Gly Lys Lys Val lie His Arg Asp lie Lys Pro Glu Asn Leu Leu Leu 195 200 205 Gly Leu Lys Gly Glu Leu Lys lie Wing Asp Phe Gly Trp Ser Val His 210 215 220 Wing Pro Ser Leu Arg Arg Lys Thr Met Cys Gly Thr Leu Asp Tyr Leu 225 230 235 240 Pro Pro Glu Met Lie Glu Gly Arg Met His Asn Glu Lys Val Asp Leu 245 250 255 Trp Cys lie Gly Val Leu Cys Tyr Glu Leu Leu Val Gly Asn Pro Pro 260 265 270 Phe Glu Ser Wing His Asn Glu Thr Tyr Arg Arg lie Val Lys Val 275 280 285 Asp Leu Lys Phe Pro Wing Ser Val Pro Thr Gly Wing Gln Asp Leu lie 290 295 300 Ser Lys Leu Leu Arg His Asn Pro Ser Glu Arg Leu Pro Leu Ala Gln 305 310 315 320 Val Ser Ala His Pro Trp Val Arg Ala Asn Ser Arg Arg Val Leu Pro 325 330 335 Pro Ser Ala Leu Gln Ser Val Ala 340 (2) INFORMATION FOR SEQ ID N0: 4 (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 403 arainoacid (B) TYPE: amino acid (C) NUMBER OF FILAMENTS: one ONLY (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: protein (iii) HYPOTHETICAL: NO (iv) SENSE CONTRARY: NO (Xi) DESCRIPTION OF THE SEQUENCE: SEQ ID N0: 4 Met Asp Arg Ser Lys Glu Asn Cys lie Ser Gly Pro Val Lys Ala Thr 1 5 10 15 Wing Pro Val Gly Gly Pro Lys Arg Val Leu Val Thr Gln Gln Phe Pro 20 25 30 Cys Gln Asn Pro Leu Pro Val Asn Ser Gly Gln Wing Gln Arg Val Leu 35 40 45 Cys Pro Ser Asn Ser Ser Gln Arg Val Pro Leu Gln Wing Gln Lys Leu 50 55 60 Val Ser Ser His Lys Pro Val Gln Asn Gln Lys Gln Lys Gln Leu Gln 65 70 75 80 Wing Thr Ser Val Pro His Pro Val Ser Arg Pro Leu Asn Asn Thr Gln 85 90 95 Lys Ser Lys Gln Pro Leu Pro Be Wing Pro Glu Asn Asn Pro Glu Glu 100 IOS 110 Glu Leu Wing Ser Lys Gln Lys Asn Glu Glu Ser Lys Lys Arg Gln Trp 115 120 125 Wing Leu Glu Asp Phe Glu lie Gly Arg Pro Leu Gly Lys Gly Lys Phe 130 135 140 Gly Asn Val Tyr Leu Wing Arg Glu Lys Gln Ser Lys Phe lie Leu Wing 145 150 155 160 Leu Lys Val Leu Phe Lys Wing Gln Leu Glu Lys Wing Gly Val Glu His 165 170 175 Gln Leu Arg Arg Glu Val Glu Lie Gln Ser His Leu Arg His Pro Asn 180 185 190 lie Leu Arg Leu Tyr Gly Tyr Phe His Asp Wing Thr Arg Val Tyr Leu 195 200 205 lie Leu Glu Tyr Ala Pro Leu Gly Thr Val Tyr Arg Glu Leu Gln Lys 210 215 220 Leu Ser Lys Phe Asp Glu Gln Arg Thr Wing Thr Tyr lie Thr Glu Leu 225 230 235 240 Wing Asn Wing Leu Ser Tyr Cys His Ser Lys Arg Val lie His Arg Asp 245 250 255 He Lys Pro Glu Asn Leu Leu Leu Gly Be Wing Gly Glu Leu Lys He 260 265 270 Wing Asp Phe Gly Trp Ser Val His Wing Pro Being Arg Arg Thr Thr 275 280 285 Leu Cys Gly Thr Leu Asp Tyr Leu Pro Pro Glu Met He Glu Gly Arg 290 295 300 Met His Asp Glu Lys Val Asp Leu Trp Ser Leu Gly Val Leu Cys Tyr 305 310 315 320 Glu Phe Leu Val Gly Lys Pro Pro Phe Glu Wing Asn Thr Tyr Gln Glu 325 330 335 Thr Tyr Lys Arg He Ser Arg Val Glu Phe Thr Phe Pro Asp Phe Val 340 345 350 Thr Glu Gly Wing Arg Asp Leu He Ser Arg Leu Leu Lys His Asn Pro 355 360 365 Ser Gln Arg Pro Met Leu Arg Glu Val Leu Glu His Pro Trp He Thr 370 375 330 Wing Asn Ser Ser Lys Pro Ser Asn Cys Gln Asn Lys Glu Ser Wing Ser 385 390 395 400 Lys Gln Ser

Claims (45)

  1. NQVEPAP PE THE INVENTION REIYINPICACIQNES 1. - An isolated or enriched purified nucleic acid molecule, characterized in that it encodes the AUR-1 and / or AUR-2 polypeptide.
  2. 2. The nucleic acid molecule according to claim 1, further characterized in that the nucleic acid is human nucleic acid.
  3. 3. The nucleic acid molecule according to claim 1, further characterized in that the molecule encodes at least 25 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID NO: 4.
  4. 4. The nucleic acid molecule according to claim 1, further characterized in that the molecule encodes at least 50 contiguous amino acids of the amino acid sequence shown in SEQ ID N0: 3 or SEQ ID N?: 4.
  5. 5. The nucleic acid molecule according to the rei indication 1, further characterized in that the molecule encodes at least 100 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID N0: 4.
  6. 6. The nucleic acid molecule according to claim 1, further characterized in that the molecule encodes at least 200 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID N?: 4.
  7. 7. The nucleic acid molecule according to claim 1, further characterized in that the molecule encodes at least 300 contiguous amino acids of the amino acid sequence shown in SEQ ID N0: 3 or SEQ ID N0: 4.
  8. 8. A nucleic acid probe for the detection of nucleic acid encoding the AUR-1 and / or AUR-2 polypeptide in a sample.
  9. 9. The probe according to claim 8, further characterized in that the polypeptide comprises at least 25 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID N0: 4.
  10. 10. The probe according to claim 8, further characterized in that the polypeptide comprises at least 50 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID NO: 4.
  11. 11. The probe according to claim 8 »further characterized in that the polypeptide comprises at least 100 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID N?: 4.
  12. 12. The probe according to claim 8 »further characterized in that the polypeptide comprises at least 200 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID N0: 4.
  13. 13. The probe according to claim 8 »further characterized in that the polypeptide comprises at least 300 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID N0: 4. 14. - A recombinant nucleic acid molecule »characterized in that it encodes an AUR-1 and / or AUR-2 polypeptide and an effective vector or promoter for initiating transcription in a host cell. 15.- A recombinant nucleic acid molecule »characterized in that it comprises a functional transcription region in a cell» a complementary sequence for an RNA sequence encoding a AUR-1 or AUR-2 polypeptide »and a transcription termination region» functional in a cell. 16.- A polypeptide AUR-l or AUR-2 isolated »enriched or purified. 17. The polypeptide AUR-1 or AUR-2 according to claim 16 »further characterized in that the polypeptide comprises at least 25 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID N0: 4 18. The polypeptide AUR-1 or AUR-2 according to claim 16 »characterized in that the polypeptide comprises at least 50 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID N?: 4. 19. The polypeptide AUR-1 or AUR-2 according to claim 16 »further characterized in that the polypeptide comprises at least 100 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID N: 4 . 20. The polypeptide AUR-1 or AUR-2 according to claim 16, further characterized in that the polypeptide comprises at least 200 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: or SEQ ID N?: 4. 21. The polypeptide AUR-1 or AUR-2 according to claim 16, further characterized in that the polypeptide comprises at least 300 contiguous amino acids of the amino acid sequence shown in SEQ ID NO: 3 or SEQ ID N?: 4. 22. The AUR-1 or AUR-2 polypeptide according to claim 16, further characterized in that the eß polypeptide isolated, purified or enriched, from a mammal. 23. The polypeptide AUR-1 or AUR-2 according to claim 16 »further characterized in that the polypeptide eß isolated, purified or enriched, from a human. 24. The AUR-1 or AUR-2 polypeptide according to claim 16, further characterized in that the polypeptide is an AUR-1 polypeptide. 25. The AUR-1 or AUR-2 polypeptide according to claim 16, further characterized in that the polypeptide is an AUR-2 polypeptide. 26.- An antibody that has a specific binding affinity for the AUR-1 or AUR-2 polypeptide. 27. The antibody according to claim 26, characterized in that the polypeptide AUR-1 or AUR-2 comprises at least 3 contiguous amino acids of the amino acid sequence shown in SEQ ID NO 3 or SEQ ID N0: 4 28.- The antibody according to the rei indication 26 »characterized in addition because the polypeptide AUR-1 or AUR-2 comprises at least 4 contiguous amino acids of the amino acid sequence shown in SEQ ID NO 3 or SEQ ID N0: 4 29. The antibody according to claim 26, characterized in that the polypeptide AUR-1 or AUR-2 comprises at least 25 contiguous amino acids of the amino acid sequence shown in SEQ ID NO 3 or SEQ ID N0: 4 30. The antibody according to claim 26 »further characterized in that the polypeptide AUR-1 or AUR-2 comprises at least 50 contiguous amino acids of the amino acid sequence shown in SEQ ID NO 3 or SEQ ID N0: 4 31. The antibody according to claim 26 »characterized in that the polypeptide AUR-1 or AUR-2 comprises at least 100 contiguous amino acids of the amino acid sequence shown in SEQ ID NO 3 or SEQ ID N0: 4 32. The antibody according to claim 26 »further characterized in that the polypeptide is isolated» purified or enriched from a mammal. 33.- The antibody according to claim 26, further characterized in that the polypeptide is isolated, purified or enriched from a human. 34. The antibody according to claim 26, further characterized in that the polypeptide is an AUR-1 polypeptide. 35. The antibody according to claim 26, further characterized in that the polypeptide is an AUR-2 polypeptide. 36.- A hybridoma, characterized in that it produces an antibody that has specific binding affinity for the AUR-1 and / or AUR-2 polypeptide. 37.- The hybridoma according to claim 36, further characterized in that the polypeptide AUR-1 or AUR-2 comprises at least 25 contiguous amino acids of the amino acid sequence shown in SEQ ID NO 3 or SEQ ID N 0: 4. 38.- The hybridoma according to claim 36, further characterized in that the polypeptide AUR-1 or AUR-2 comprises at least 50 contiguous amino acids of the amino acid sequence shown in SEQ ID NO 3 or SEQ ID N 4: 4. 39. The hybridoma according to the rei indication 36, further characterized in that the AUR-1 or AUR-2 polypeptide comprises at least 100 contiguous amino acids of the amino acid sequence shown in SEQ ID NO 3 or SEQ ID N: Four. 40. The hybridoma according to claim 36, characterized in that the AUR-1 or AUR-2 polypeptide comprises at least 200 contiguous inoacids of the amino acid sequence shown in SEQ ID NO.3 or SEQ ID N ?: Four. 41. The hybridizer according to claim 36, further characterized in that the polypeptide AUR-1 or AUR-2 comprises? at least 300 contiguous amino acids of the amino acid sequence shown in SEQ ID NO.3 or SEQ ID N: 4. 42.- The hybridoma according to claim 36, characterized in addition to the isolated eß polypeptide, purified or enriched from a mammal. 43. The hybridoma according to claim 36, further characterized in that the polypeptide is isolated, purified or enriched from a human. 44. The hybridoma according to claim 36, further characterized in that the polypeptide is an AUR-1 polypeptide. 45.- The hybridoma according to the rei indication 36, further characterized in that the polypeptide is an AUR-2 polypeptide.
MXPA/A/1998/004934A 1995-12-18 1998-06-18 Diagnosis and treatment of alterations related to aur-1 and / or au MXPA98004934A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US008809 1995-12-18
US023943 1996-08-14

Publications (1)

Publication Number Publication Date
MXPA98004934A true MXPA98004934A (en) 1999-04-06

Family

ID=

Similar Documents

Publication Publication Date Title
AU716330B2 (en) Diagnosis and treatment of AUR-1 and/or AUR-2 related disorders
EP1051500B1 (en) Diagnosis and treatment of aur1 and/or aur2 related disorders
EP1073723B1 (en) Ste20-related protein kinases
US20060292573A1 (en) Human orthologues of WART
US20080009610A1 (en) Diagnosis and treatment of PTP related disorders
US7119174B2 (en) Diagnosis and treatment of AUR1 and/or AUR2 related disorders
EP0942934A2 (en) Receptor tyrosine kinase genes
US6844177B2 (en) Diagnosis and treatment of PTP04 related disorders
MXPA98004934A (en) Diagnosis and treatment of alterations related to aur-1 and / or au
US6342593B1 (en) Diagnosis and treatment of ALP related disorders